US20080167339A1 - Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors - Google Patents

Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors Download PDF

Info

Publication number
US20080167339A1
US20080167339A1 US11/950,932 US95093207A US2008167339A1 US 20080167339 A1 US20080167339 A1 US 20080167339A1 US 95093207 A US95093207 A US 95093207A US 2008167339 A1 US2008167339 A1 US 2008167339A1
Authority
US
United States
Prior art keywords
formula
alkyl
alkylene
pharmaceutically acceptable
hydrogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/950,932
Other versions
US7846943B2 (en
Inventor
Gerhard Zoller
Stefan Petry
Gunter Muller
Hubert Heuer
Norbert Tennagels
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi SA
Original Assignee
Sanofi Aventis France
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi Aventis France filed Critical Sanofi Aventis France
Assigned to SANOFI-AVENTIS reassignment SANOFI-AVENTIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HEUER, HUBERT, TENNAGELS, NORBERT, MULLER, GUNTER, PETRY, STEFAN, ZOLLER, GERHARD
Publication of US20080167339A1 publication Critical patent/US20080167339A1/en
Priority to US12/914,045 priority Critical patent/US20110039883A1/en
Application granted granted Critical
Publication of US7846943B2 publication Critical patent/US7846943B2/en
Assigned to SANOFI reassignment SANOFI CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SANOFI-AVENTIS
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/424Oxazoles condensed with heterocyclic ring systems, e.g. clavulanic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention relates to azolopyridin-2-one derivatives of the general formula I, to their pharmaceutically useful salts and to their use as medicinal substances.
  • the present invention provides compounds which have an inhibitory effect on hormone-sensitive lipase or endothelial lipase.
  • the invention relates to azolopyridin-2-one derivatives of the general formula I
  • Preferred compounds of the formula I are those in which
  • Preferred compounds of the formula I are also those in which
  • the invention relates to compounds of the formula I in the form of their salts, racemates, racemic mixtures and pure enantiomers, and to their diastereomers and mixtures thereof.
  • alkyl and alkylene radicals in the substituents R, R1, R2, R3, R4, R5, R6 and R7 may be either straight-chain or branched.
  • Halogen is fluorine, chlorine, bromine or iodine, in particular fluorine or chlorine.
  • the alkyl and alkylene radicals may be substituted one or more times by suitable groups
  • Aryl means an aromatic carbocyclic mono- or bicyclic ring system which comprises 6 to 10 atoms in the ring or in the rings.
  • the aryl radicals may be substituted one or more times by suitable groups.
  • Heteroaryl is a mono- or bicyclic aromatic ring system having 5 to 12 ring members, in which at least one atom in the ring system is a heteroatom from the series N, O and S.
  • Suitable “heteroaryl rings” or “heteroaryl radicals” are, for example, benzimidazolyl, benzofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, quinolinyl, furyl, furazanyl, imidazolyl, 1H-indazolyl, indolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyridyl, pyrrolyl, thiazolyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl,
  • Preferred heteroaryl radicals are thiophenyl, pyrazolyl and pyridyl.
  • the heteroaryl rings or heteroaryl radicals may be substituted one or more times by suitable groups.
  • a cycloalkyl radical is a ring system which comprises one or more rings and which is in saturated or partially unsaturated (with one or two double bonds) form and which is composed exclusively of carbon atoms.
  • the cycloalkyl radicals may be substituted one or more times by suitable groups.
  • Bicycle means bicyclic ring systems which are in saturated or partially unsaturated form and which, apart from carbon, may also comprise one or more heteroatoms such as, for example, nitrogen, oxygen or sulfur.
  • This definition also includes ring systems which comprise a fused benzene nucleus. Examples which may be mentioned are the tetrahydronaphthyl, alpha- or beta-tetralon-, indanyl- or indan-1-on-yl radical.
  • Preferred bicyclic radicals are those of formula Ic
  • bicyclic radicals may be substituted one or more times by suitable groups.
  • Suitable pharmaceutically acceptable acid addition salts of the compounds of the invention are salts of inorganic acids such as hydrochloric acid, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acid, and of organic acids such as, for example, acetic acid, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isethionic, lactic, lactobionic, maleic, malic, methanesulfonic, succinic, p-toluenesulfonic and tartaric acid.
  • inorganic acids such as hydrochloric acid, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acid
  • organic acids such as, for example, acetic acid, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isethionic, lactic, lactobionic, maleic
  • Suitable pharmaceutically acceptable basic salts are ammonium salts, alkali metal salts (such as sodium and potassium salts) and alkaline earth metal salts (such as magnesium and calcium salts) and salts of trometamol (2-amino-2-hydroxymethyl-1,3-propanediol), diethanolamine, lysine or ethylenediamine.
  • Salts with a pharmaceutically unacceptable anion such as, for example, trifluoroacetate likewise belong within the framework of the invention as useful intermediates for the preparation or purification of pharmaceutically acceptable salts and/or for use in nontherapeutic, for example in vitro, applications.
  • physiologically functional derivative refers to any physiologically tolerated derivative of a compound of the invention of the formula I, for example an ester, which on administration to a mammal such as, for example, a human is able to form (directly or indirectly) a compound of the formula I or an active metabolite thereof.
  • Physiologically functional derivatives also include prodrugs of the compounds of the invention as, for example, described in H. Okada et al., Chem. Pharm. Bull. 1994, 42, 57-61. Such prodrugs can be metabolized in vivo to a compound of the invention. These prodrugs may themselves be active or not.
  • the compounds of the invention may also exist in various polymorphous forms, for example as amorphous and crystalline polymorphous forms. All polymorphous forms of the compounds of the invention belong within the framework of the invention and are a further aspect of the invention.
  • the compounds of the invention of the general formula I have a surprising inhibitory effect on hormone sensitive lipase, HSL, an allosteric enzyme in adipocytes which is inhibited by insulin and is responsible for the breakdown of fats in fat cells and thus for transferring fat constituents into the blood stream. Inhibition of this enzyme is therefore equivalent to an insulin-like effect of the compounds of the invention, eventually leading to a reduction of free fatty acids in the blood and of blood glucose. They can therefore be employed for metabolic derangements such as, for example, for non-insulin-dependent diabetes mellitus, for diabetic syndrome and for direct pancreatic damage.
  • the compounds of the invention of the general formula I may additionally have an inhibitory effect on endothelial lipase (EL).
  • EL endothelial lipase
  • the preferred substrate for EL is HDL, which has antiatherosclerotic activity. A reduction in the HDL level leads to progression of atherosclerosis and its sequelae such as coronary heart disease and moreover favors development of the metabolic syndrome and its sequelae diabetes. An inhibition of EL should thus generally lead to prevention of atherosclerotic disorders and indirectly reduce the probability of people with an increased risk for diabetes becoming ill.
  • the compounds of the invention of the formula I may also have an inhibitory effect on triglyceride lipase.
  • Compounds of this type are particularly suitable for the treatment and/or prevention of
  • the amount of a compound of the invention necessary to achieve the desired biological effect depends on a number of factors, for example the specific compound chosen, the intended use, the mode of administration and the clinical condition of the patient.
  • the daily dose is generally in the range from 0.3 mg to 100 mg (typically from 3 mg to 50 mg) per day and per kilogram of body weight, for example 3-10 mg/kg/day.
  • An intravenous dose may be, for example, in the range from 0.3 mg to 1.0 mg/kg, which can suitably be administered as infusion of 10 ng to 100 ng per kilogram and per minute.
  • Suitable infusion solutions for these purposes may contain, for example, from 0.1 ng to 10 mg, typically from 1 ng to 10 mg, per milliliter.
  • Single doses may contain, for example, from 1 mg to 10 g of the active ingredient.
  • ampoules for injections may contain, for example, from 1 mg to 100 mg
  • single-dose formulations which can be administered orally, such as, for example, tablets or capsules may contain, for example, from 0.05 to 1000 mg, typically from 0.5 to 600 mg.
  • the compounds of formula I may be used as the compound itself, but they are preferably in the form of a pharmaceutical composition with an acceptable carrier.
  • the carrier must, of course, be acceptable in the sense that it is compatible with the other ingredients of the composition and is not harmful for the patient's health.
  • the carrier may be a solid or a liquid or both and is preferably formulated with the compound as a single dose, for example as a tablet, which may contain from 0.05% to 95% by weight of the active ingredient.
  • Other pharmaceutically active substances may likewise be present, including other compounds of the invention.
  • the pharmaceutical compositions of the invention can be produced by one of the known pharmaceutical methods, which essentially consist of mixing the ingredients with pharmacologically acceptable carriers and/or excipients.
  • compositions of the invention are those suitable for oral, rectal, topical, peroral (for example sublingual) and parenteral (for example subcutaneous, intramuscular, intradermal or intravenous) administration, although the most suitable mode of administration depends in each individual case on the nature and severity of the condition to be treated and on the nature of the compound of formula I used in each case.
  • Coated formulations and coated slow-release formulations also belong within the framework of the invention. Preference is given to acid- and gastric juice-resistant formulations. Suitable coatings resistant to gastric juice comprise cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropylmethylcellulose phthalate and anionic polymers of methacrylic acid and methyl methacrylate.
  • Suitable pharmaceutical preparations for oral administration may be in the form of separate units such as, for example, capsules, cachets, suckable tablets or tablets, each of which contain a defined amount of the compound of formula I; as powders or granules; as solution or suspension in an aqueous or nonaqueous liquid; or as an oil-in-water or water-in-oil emulsion.
  • These compositions may, as already mentioned, be prepared by any suitable pharmaceutical method which includes a step in which the active ingredient and the carrier (which may consist of one or more additional ingredients) are brought into contact.
  • the compositions are generally produced by uniform and homogeneous mixing of the active ingredient with a liquid and/or finely divided solid carrier, after which the product is shaped if necessary.
  • a tablet can be produced by compressing or molding a powder or granules of the compound, where appropriate with one or more additional ingredients.
  • Compressed tablets can be produced by tableting the compound in free-flowing form such as, for example, a powder or granules, where appropriate mixed with a binder, glidant, inert diluent and/or one or more surface-active/dispersing agent(s) in a suitable machine.
  • Molded tablets can be produced by molding the compound, which is in powder form and is moistened with an inert liquid diluent, in a suitable machine.
  • compositions which are suitable for peroral (sublingual) administration comprise suckable tablets which contain a compound of formula I with a flavoring, normally sucrose and gum arabic or tragacanth, and pastilles which comprise the compound in an inert base such as gelatin and glycerol or sucrose and gum arabic.
  • compositions suitable for parenteral administration comprise preferably sterile aqueous preparations of a compound of formula I, which are preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although administration may also take place by subcutaneous, intramuscular or intradermal injection. These preparations can preferably be produced by mixing the compound with water and making the resulting solution sterile and isotonic with blood. Injectable compositions of the invention generally contain from 0.1 to 5% by weight of the active compound.
  • compositions suitable for rectal administration are preferably in the form of single-dose suppositories. These can be produced by mixing a compound of the formula I with one or more conventional solid carriers, for example cocoa butter, and shaping the resulting mixture.
  • compositions suitable for topical use on the skin are preferably in the form of ointment, cream, lotion, paste, spray, aerosol or oil.
  • Carriers which can be used are petrolatum, lanolin, polyethylene glycols, alcohols and combinations of two or more of these substances.
  • the active ingredient is generally present in a concentration of from 0.1 to 15% by weight of the composition, for example from 0.5 to 2%.
  • compositions suitable for transdermal uses can be in the form of single patches which are suitable for long-term close contact with the patient's epidermis. Such patches suitably contain the active ingredient in an aqueous solution which is buffered where appropriate, dissolved and/or dispersed in an adhesive or dispersed in a polymer.
  • a suitable active ingredient concentration is about 1% to 35%, preferably about 3% to 15%.
  • a particular possibility is for the active ingredient to be released by electrotransport or iontophoresis as described, for example, in Pharmaceutical Research, 2(6): 318 (1986).
  • the compounds of the formula I with R2 hydrogen are distinguished by favorable effects on disorders of lipid metabolism. They beneficially influence the HDL to LDL ratio and increase in particular the HDL level and are suitable for the prevention and treatment of dyslipidemias and metabolic syndrome and their diverse sequelae such as atherosclerosis, coronary heart disease, heart failure, obesity and diabetes.
  • the compounds of the formula I with R1 and R2, which form a ring system, are distinguished by favorable effects on metabolic disorders. They beneficially influence lipid and sugar metabolism, in particular they lower the triglyceride level and are suitable for the prevention and treatment of type II diabetes and arteriosclerosis and the diverse sequelae thereof.
  • the compounds of the invention can be administered alone or in combination with one or more further pharmacologically active substances which have, for example, favorable effects on metabolic disturbances or disorders frequently associated therewith.
  • further pharmacologically active substances which have, for example, favorable effects on metabolic disturbances or disorders frequently associated therewith. Examples of such medicaments are
  • Administration of the active ingredient combination can take place either by separate administration of the active ingredients to the patient or in the form of combination products in which a plurality of active ingredients are present in one pharmaceutical preparation.
  • Antidiabetics include all insulins and insulin derivatives such as, for example, Lantus® (see www.lantus.com) or Apidra®, and other fast-acting insulins (see U.S. Pat. No. 6,221,633), GLP-1 receptor modulators as described in WO 01/04146 or else, for example, those disclosed in WO 98/08871 of Novo Nordisk A/S.
  • the orally effective hypoglycemic active ingredients include, preferably, the sulfonylureas which act on the ATP-dependent potassium channel of the beta cells (e.g. disclosed in WO 97/26265 and WO 99/03861), biguanides, meglitinides, glucagon antagonists, oral GLP-1 agonists, DPP-IV inhibitors, insulin sensitizers, e.g.
  • PPAR and PXR modulators and active ingredients such as, for example, oxadiazolidinediones, thiazolidinediones, inhibitors of liver enzymes which are involved in stimulating gluconeogenesis and/or glycogenolysis, modulators of glucose uptake such as, for example, glucosidase inhibitors, compounds which alter lipid metabolism and lead to a change in the blood lipid composition, compounds which reduce food intake or food uptake.
  • the compounds of the formula I are administered in combination with insulin.
  • the compounds of the formula I are administered in combination with substances which influence hepatic glucose production such as, for example, glycogen phosphorylase inhibitors (see: WO 01/94300, WO 02/096864, WO 03/084923, WO 03/084922, WO 03/104188).
  • substances which influence hepatic glucose production such as, for example, glycogen phosphorylase inhibitors (see: WO 01/94300, WO 02/096864, WO 03/084923, WO 03/084922, WO 03/104188).
  • the compounds of the formula I are administered in combination with an active ingredient which acts on the ATP-dependent potassium channel of the beta cells, such as, for example, sulfonylureas (e.g. tolbutamide, glibenclamide, glipizide, glimepiride) or glinides (e.g. repaglinide).
  • an active ingredient which acts on the ATP-dependent potassium channel of the beta cells, such as, for example, sulfonylureas (e.g. tolbutamide, glibenclamide, glipizide, glimepiride) or glinides (e.g. repaglinide).
  • the compounds of the formula I are administered in combination with a biguanide such as, for example, metformin.
  • a biguanide such as, for example, metformin.
  • the compounds of the formula I are administered in combination with a PPARgamma agonist or thiazolidinedione such as, for example, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed in WO 97/41097 of Dr. Reddy's Research Foundation, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2-quinazolinylmethoxy]phenyl]methyl]-2,4-thiazolidinedione.
  • a PPARgamma agonist or thiazolidinedione such as, for example, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed in WO 97/41097 of Dr. Reddy's Research Foundation, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2-quinazolinylmethoxy]phenyl]methyl]-2,4-thi
  • the compounds of the formula I are administered in combination with a DPPIV inhibitor as described, for example, in WO98/19998, WO99/61431, WO99/67278, WO99/67279, WO01/72290, WO 02/38541, WO03/040174, in particular P 93/01 (1-cyclopentyl-3-methyl-1-oxo-2-pentanammonium chloride), P-31/98, LAF237 (1-[2-[3-hydroxyadamant-1-ylamino)acetyl]pyrrolidine-2-(S)-carbonitrile), TS021 ((2S,4S)-4-fluoro-1-[[(2-hydroxy-1,1-dimethylethyl)amino]-acetyl]pyrrolidine-2-carbonitrile monobenzenesulfonate).
  • a DPPIV inhibitor as described, for example, in WO98/19998, WO99/61431, WO99/67278, WO99/672
  • the compounds of the formula I are administered in combination with compounds with an inhibitory effect on SGLT-1 and/or 2, as disclosed directly or indirectly for example in WO2004/007517, WO2004/052902, WO2004/052903 and WO2005/121161.
  • the compounds of the formula I are administered in combination with an ⁇ -glucosidase inhibitor such as, for example, miglitol or acarbose.
  • an ⁇ -glucosidase inhibitor such as, for example, miglitol or acarbose.
  • the compounds of the formula I are administered in combination with more than one of the aforementioned compounds, e.g. in combination with a sulfonylurea and metformin, a sulfonylurea and acarbose, repaglinide and metformin, insulin and a sulfonylurea, insulin and metformin, insulin and troglitazone, insulin and lovastatin, etc.
  • the compounds of the formula I are administered in combination with an HMGCoA reductase inhibitor such as lovastatin, fluvastatin, pravastatin, simvastatin, ivastatin, itavastatin, atorvastatin, rosuvastatin.
  • an HMGCoA reductase inhibitor such as lovastatin, fluvastatin, pravastatin, simvastatin, ivastatin, itavastatin, atorvastatin, rosuvastatin.
  • the compounds of the formula I are administered in combination with a bile acid absorption inhibitor (see, for example, U.S. Pat. No. 6,245,744, U.S. Pat. No. 6,221,897, U.S. Pat. No. 6,277,831, EP 0683 773, EP 0683 774).
  • a bile acid absorption inhibitor see, for example, U.S. Pat. No. 6,245,744, U.S. Pat. No. 6,221,897, U.S. Pat. No. 6,277,831, EP 0683 773, EP 0683 774.
  • the compounds of the formula I are administered in combination with a polymeric bile acid adsorbent such as, for example, cholestyramine, colesevelam.
  • a polymeric bile acid adsorbent such as, for example, cholestyramine, colesevelam.
  • the compounds of the formula I are administered in combination with a cholesterol resorption inhibitor as described for example in WO 0250027, or ezetimibe, tiqueside, pamaqueside.
  • the compounds of the formula I are administered in combination with an LDL receptor inducer (see, for example, U.S. Pat. No. 6,342,512).
  • the compounds of the formula I are administered in combination with bulking agents, preferably insoluble bulking agents (see, for example, carob/Caromax® (Zunft H J; et al., Carob pulp preparation for treatment of hypercholesterolemia, ADVANCES IN THERAPY (2001 September-October), 18(5), 230-6)).
  • Caromax is a carob-containing product from Nutrinova, Nutrition Specialties & Food Ingredients GmbH, Industriepark Hoechst, 65926 Frankfurt/Main).
  • Combination with Caromax® is possible in one preparation or by separate administration of compounds of the formula I and Caromax®.
  • Caromax® can in this connection also be administered in the form of food products such as, for example, in bakery products or muesli bars.
  • the compounds of the formula I are administered in combination with a PPARalpha agonist.
  • the compounds of the formula I are administered in combination with a mixed PPAR alpha/gamma agonist such as, for example, AZ 242 (tesaglitazar, (S)-3-(4-[2-(4-methanesulfonyloxyphenyl)ethoxy]-phenyl)-2-ethoxypropionic acid), BMS 298585 (N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine) or as described in WO 99/62872, WO 99/62871, WO 01/40171, WO 01/40169, WO96/38428, WO 01/81327, WO 01/21602, WO 03/020269, WO 00/64888 or WO 00/64876.
  • AZ 242 tesaglitazar, (S)-3-(4-[2-(4-
  • the compounds of the formula I are administered in combination with a fibrate such as, for example, fenofibrate, gemfibrozil, clofibrate, bezafibrate.
  • a fibrate such as, for example, fenofibrate, gemfibrozil, clofibrate, bezafibrate.
  • the compounds of the formula I are administered in combination with nicotinic acid or niacin.
  • the compounds of the formula I are administered in combination with a CETP inhibitor, e.g. CP-529, 414 (torcetrapib).
  • a CETP inhibitor e.g. CP-529, 414 (torcetrapib).
  • the compounds of the formula I are administered in combination with an ACAT inhibitor.
  • the compounds of the formula I are administered in combination with an MTP inhibitor such as, for example, implitapide.
  • the compounds of the formula I are administered in combination with an antioxidant.
  • the compounds of the formula I are administered in combination with a lipoprotein lipase inhibitor.
  • the compounds of the formula I are administered in combination with an ATP citrate lyase inhibitor.
  • the compounds of the formula I are administered in combination with a squalene synthetase inhibitor.
  • the compounds of the formula I are administered in combination with a lipoprotein(a) antagonist.
  • the compounds of the formula I are administered in combination with a lipase inhibitor such as, for example, orlistat.
  • the further active ingredient is fenfluramine or dexfenfluramine. In another embodiment, the further active ingredient is sibutramine.
  • the compounds of the formula I are administered in combination with CART modulators (see “Cocaine-amphetamine-regulated transcript influences energy metabolism, anxiety and gastric emptying in mice” Asakawa, A, et al., M.: Hormone and Metabolic Research (2001), 33(9), 554-558), NPY antagonists, e.g. naphthalene-1-sulfonic acid ⁇ 4-[(4-aminoquinazolin-2-ylamino)methyl]cyclohexylmethyl ⁇ amide; hydrochloride (CG P 71683A)), MC4 agonists (e.g.
  • CRF BP antagonists e.g. urocortin
  • urocortin agonists e.g. urocortin
  • urocortin agonists e.g. urocortin
  • urocortin agonists e.g. urocortin agonists
  • ⁇ 3 agonists e.g. 1-(4-chloro-3-methanesulfonylmethylphenyl)-2-[2-(2,3-dimethyl-1H-indol-6-yloxy)ethylamino]ethanol; hydrochloride (WO 01/83451)
  • MSH melanocyte-stimulating hormone
  • CCK-A agonists e.g.
  • 1-(3-ethylbenzofuran-7-yl)piperazine oxalic acid salt (WO 01/09111), bombesin agonists, galanin antagonists, growth hormone (e.g. human growth hormone), growth hormone-releasing compounds (6-benzyloxy-1-(2-diisopropylaminoethylcarbamoyl)-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tertiary butyl ester (WO 01/85695)), TRH agonists (see, for example, EP 0 462 884), uncoupling protein 2 or 3 modulators, leptin agonists (see, for example, Lee, Daniel W.; Leinung, Matthew C.; Rozhavskaya-Arena, Marina; Grasso, Patricia.
  • growth hormone e.g. human growth hormone
  • growth hormone-releasing compounds (6-benzyloxy-1-(2-diisopropylaminoethylcar
  • Leptin agonists as a potential approach to the treatment of obesity. Drugs of the Future (2001), 26(9), 873-881), DA agonists (bromocriptine, Doprexin), lipase/amylase inhibitors (e.g. WO 00/40569), PPAR modulators (e.g. WO 00/78312), RXR modulators or TR- ⁇ agonists.
  • DA agonists bromocriptine, Doprexin
  • lipase/amylase inhibitors e.g. WO 00/40569
  • PPAR modulators e.g. WO 00/78312
  • RXR modulators or TR- ⁇ agonists e.g. WO 00/78312
  • the further active ingredient is a cannabinoid receptor 1 antagonist (such as, for example, rimonabant, SR147778 or those as are described in, for example, EP 0656354, WO 00/15609, WO 02/076949, WO2005080345, WO2005080328, WO2005/080343, WO2005/075450, WO2005/080357, WO2001/70700, WO2003/026647-48, WO2003/02776, WO2003/040107, WO2003/007887, WO2003/027069, U.S. Pat. No.
  • a cannabinoid receptor 1 antagonist such as, for example, rimonabant, SR147778 or those as are described in, for example, EP 0656354, WO 00/15609, WO 02/076949, WO2005080345, WO2005080328, WO2005/080343, WO2005/075450, WO2005/08
  • the further active ingredient is leptin.
  • the further active ingredient is dexamphetamine, amphetamine, mazindole or phentermine.
  • the compounds of the formula I are administered in combination with medicaments having effects on the coronary circulation and the vascular system, such as, for example, ACE inhibitors (e.g. ramipril), medicaments which act on the angiotensin-renine system, calcium antagonists, beta blockers etc.
  • medicaments having effects on the coronary circulation and the vascular system such as, for example, ACE inhibitors (e.g. ramipril), medicaments which act on the angiotensin-renine system, calcium antagonists, beta blockers etc.
  • the compounds of the formula I are administered in combination with medicaments having an antiinflammatory effect.
  • the compounds of the formula I are administered in combination with medicaments which are employed for cancer therapy and cancer prevention.
  • Isolated rat fat cells are obtained from epididymal adipose tissue from untreated male rats (Wistar, 220-250 g) by collagenase treatment in accordance with published methods (e.g. S, Nilsson et al., Anal. Biochem. 158, 1986, 399-407; G. Fredrikson et al., J. Biol. Chem. 256, 1981, 6311-6320; H. Tornquist et al., J. Biol. Chem. 251, 1976, 813-819).
  • the fat cells from 10 rats are washed three times by flotation with 50 ml of homogenization buffer (25 ml Tris/HCl, pH 7.4, 0.25 M sucrose, 1 mM ETDA, 1 mM DTT, 10 ⁇ g/ml leupeptin, 10 ⁇ g/ml antipain, 20 ⁇ g/ml pepstatin) each time and finally taken up in 10 ml of homogenization buffer.
  • the fat cells are homogenized in a Teflon-in-glass homogenizer (Braun-Melsungen) by 10 strokes at 1500 rpm and 15° C.
  • the homogenate is centrifuged (Sorvall SM24 tubes, 5000 rpm, 10 min, 4° C.).
  • the subnatant between the layer of fat at the top and the pellet is removed and the centrifugation is repeated.
  • the subnatant resulting therefrom is centrifuged again (Sorvall SM24 tubes, 20 000 rpm, 45 min, 4° C.).
  • the subnatant is removed, and 1 g of heparin-Sepharose (Pharmacia-Biotech, CL-6B, washed 5 ⁇ with 25 mM Tris/HCl, pH 7.4, 150 mM NaCl) is added. After incubation at 4° C. for 60 min (shaking at intervals of 15 min), the mixture is centrifuged (Sorvall SM24 tubes, 3000 rpm, 10 min, 4° C.).
  • the supernatant is adjusted to pH 5.2 by adding glacial acetic acid and is incubated at 4° C. for 30 min.
  • the precipitates are collected by centrifugation (Sorvall SS34, 12 000 rpm, 10 min, 4° C.) and suspended in 2.5 ml of 20 mM Tris/HCl, pH 7.0, 1 mM EDTA, 65 mM NaCl, 13% sucrose, 1 mM DTT, 10 ⁇ g/ml leupeptin/pepstatin/antipain.
  • the suspension is dialyzed against 25 mM Tris/HCl, pH 7.4, 50% glycerol, 1 mM DTT, 10 ⁇ g/ml leupeptin, pepstatin, antipain at 4° C. overnight and then loaded onto a hydroxiapatite column (0.1 g per 1 ml of suspension, equilibrated with 10 mM potassium phosphate, pH 7.0, 30% glycerol, 1 mM DTT). The column is washed with four volumes of equilibration buffer at a flow rate of 20 to 30 ml/h.
  • the HSL is eluted with one volume of equilibration buffer containing 0.5 M potassium phosphate and then dialyzed (see above) and concentrated 5- to 10-fold by ultrafiltration (Amicon Diaflo PM 10 Filter) at 4° C.
  • the partially purified HSL can be stored at ⁇ 70° C. for 4 to 6 weeks.
  • Substances are normally tested in four independent mixtures.
  • the inhibition of the HSL enzymatic activity by a test substance is determined by comparing with an uninhibited control reaction.
  • the IC 50 is calculated from an inhibition plot with at least 10 concentrations of the test substance.
  • the GRAPHIT, Elsevier-BIOSOFT software package is used to analyze the data.
  • EL is released as secretory protein in high concentration into cell culture medium (conditioned medium) by recombinant cell lines (CHO, HEK293). This is employed as enzyme solution after concentration.
  • the phospholipase-specific substrate 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phosphocholine, (manufacturer Molecular Probes) is used to characterize the enzymatic activity of endothelial lipase and the effect of inhibitors. Hydrolysis of the A1 ester linkage of this phospholipid by the enzyme liberates the fluorescent dye Bodipy which can be detected after separation by thin-layer chromatography on an HPTLC plate (silica gel 60, Merck) or directly in the reaction vessel by measuring the fluorescence.
  • the substrate solution is prepared by taking up 100 ⁇ g of 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phospho-choline (manufacturer Molecular Probes), 2.4 mg of tripalmitin (Sigma) and 7.9 mg of DOP-choline (1,2-dioleoyl-sn-glycero-3-phosphocholine) in 393 ⁇ l of chloroform and then transferring 157 ⁇ l into a fresh reaction vessel.
  • 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phospho-choline manufactured by taking up 100 ⁇ g of 1,2-bis(4,4-difluoro-5,7-dimethyl-4-
  • the subsequent enzymic reaction takes place at 37° C. for 60 minutes.
  • 45 ⁇ l of the substrate solution are incubated with 1 ⁇ l of inhibitor of appropriate concentration (dissolved in DMSO, pure DMSO solution is used as control) and 5 ⁇ l of enzyme solution (conditioned medium).
  • the enzymatic activity is reduced as a function of the inhibitor concentration used, and the inhibitor concentration at which a half-maximum enzymatic activity is observed is called IC 50 .
  • the phospholipase-specific substrate 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phosphocholine, (manufacturer Molecular Probes) is used to characterize the enzymatic activity of endothelial lipase and the effect of inhibitors. Hydrolysis of the A1 ester linkage of this phospholipid by the enzyme liberates the fluorescent dye Bodipy which can be detected after separation by thin-layer chromatography on an HPTLC plate (silica gel 60, Merck) or directly in the reaction vessel by measuring the fluorescence.
  • the substrate solution is prepared by dissolving 100 ⁇ g of 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phospho-choline (manufacturer Molecular Probes) in 100 ⁇ l of DMSO and taking up in 2.4 mg of tripalmitin (Sigma) in 393 ⁇ l of chloroform which comprises 20 mg/ml DOP-choline (1,2-dioleoyl-sn-glycero-3-phosphocholine). 39.3 ⁇ l of this lipid mixture are transferred into a fresh reaction vessel, and the solvent is evaporated off.
  • the subsequent enzymic reaction takes place at 37° C. for 90 minutes.
  • 20 ⁇ l of the substrate solution are incubated with 2 ⁇ l of inhibitor of appropriate concentration (dissolved in 10% DMSO, using 10% strength DMSO solution for control) and 2 ⁇ l of enzyme solution (conditioned medium).
  • the 4 ⁇ l of the assay mixture are loaded onto an HPTLC plate (silica gel 60, Merck), and the liberated fluorescent dye is separated for detection with an eluent (diethyl ether:petroleum benzine:acetic acid [78:22:1]). After evaporation of the eluent, the plate is read in a fluorescence scanner. An increased liberation of the fluorescent dye in the uninhibited reaction is to be observed as a measure of the enzymic activity.
  • the compounds of the invention of the general formula I are prepared by methods known per se, e.g. by acylation of substituted or unsubstituted azolopyridin-2-one derivatives II with carbamoyl chlorides III (method A), or in two stages by reacting azolopyridin-2-one derivatives II with phosgene or equivalents such as trichloromethyl chlorocarbonate, ditrichloromethyl carbonate or 4-nitrophenyl chloroformate and further reaction of the resulting azolopyridin-2-onecarboxylic acid derivative with amines IV (method B).
  • R2 is hydrogen
  • the azolopyridin-2-one derivatives II can also be reacted with the appropriate isocyanates V R1-N ⁇ C ⁇ O.
  • bases such as pyridine, triethylamine, sodium hydroxide solution or alkali metal carbonates for expedition.
  • the reactions can be carried out in wide temperature ranges. It has usually proved to be advantageous to operate at from 0° C. to the boiling point of the solvent used.
  • solvents employed are methylene chloride, THF, DMF, toluene, ethyl acetate, n-heptane, dioxane, diethyl ether or pyridine. If anhydrous conditions are used, strong bases such as lithium hydride, sodium hydride or potassium tert-butoxide in aprotic solvents such as THF or DMF have also proved suitable.
  • azolopyridin-2-one derivatives or corresponding aza-substituted derivatives employed as starting compounds II are commercially available or can be prepared by processes known from the literature (e.g. C. Flouzat, Y. Bresson, A. Mattio, J. Bonnet, G. Nicolast J. Med. Chem. 1993, 36, 497-503; F. Mutterer, C. D. Weis, J. Het. Chem. 1976, 13, 1103-1104; K. Bowden, G. Crank, W. J. Ross, J. Chem. Soc. 1968, 172-185).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Diabetes (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Emergency Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention provides compounds which have an inhibitory effect on hormone-sensitive lipase or endothelial lipase. The compounds of the invention are azolopyridin-2-one derivatives of the general formula I
Figure US20080167339A1-20080710-C00001
wherein the substituents are as defined herein. The compounds of formula I wherein R2 is hydrogen are distinguished by favorable effects on disorders of lipid metabolism. They beneficially influence the HDL to LDL ratio and increase in particular the HDL level and are suitable for the prevention and treatment of dyslipidemias and metabolic syndrome and their diverse sequelae such as atherosclerosis, coronary heart disease, heart failure, obesity and diabetes. The compounds of formula I wherein R1 and R2 together form a ring system are distinguished by favorable effects on metabolic disorders. They beneficially influence lipid and sugar metabolism; in particular they lower the triglyceride level and are suitable for the prevention and treatment of type II diabetes and arteriosclerosis and the diverse sequelae thereof.

Description

  • This application is a continuation of International application No. PCT/EP2006/005,095, filed May 27, 2006, which is incorporated herein by reference in its entirety; which claims the benefit of priority of German Patent Application No. 10 2005 026 762.9, filed Jun. 9, 2005.
  • The present invention relates to azolopyridin-2-one derivatives of the general formula I, to their pharmaceutically useful salts and to their use as medicinal substances.
  • Compounds of similar structure are known from EP 1275653. However, these are used as fungicides.
  • Compounds having an inhibitory effect on hormone-sensitive lipase are described in the prior art for example in WO2004/035550, WO2005/073199 or WO03/051842. Compounds with an inhibitory effect on endothelial lipase are described in the prior art for example in WO2004/094394, WO2004/094393.
  • The present invention provides compounds which have an inhibitory effect on hormone-sensitive lipase or endothelial lipase.
  • The invention relates to azolopyridin-2-one derivatives of the general formula I
  • Figure US20080167339A1-20080710-C00002
  • in which the meanings are:
    • X identically or differently ═C(—R)— or ═N—, where at least one and at most two ═C(—R—)— is replaced by ═N—;
    • Y —O— or —S—;
    • R identically or differently hydrogen, halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy-(C1-C3)-alkylene, hydroxy, (C1-C6)-alkylmercapto, amino, (C1-C6)-alkylamino, di-(C2-C12)-alkylamino, (C1-C6)-alkylcarbonyl, COOR3, trifluoromethyl, (C1-C6)-alkylsulfonyl, (C1-C6)-alkylsulfinyl, aminosulfonyl, pentafluorosulfanyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, CO—NR6R7, O—CO—NR6R7, O—CO—(C1-C6)-alkylene-CO—O—(C1-C6)-alkyl, O—CO—(C1-C6)-alkylene-CO—OH, O—CO—(C1-C6)-alkylene-CO—NR6R7 or unsubstituted or mono- or poly-F-substituted (C1-C6)-alkyloxy;
    • R1 is (C4-C16)-alkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C3-C12)-cycloalkyl, (C8-C14)-bicycle, where aryl, heteroaryl, cycloalkyl or bicycle may be substituted one or more times by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, (C1-C6)-alkylmercapto, amino, (C1-C6)-alkylamino, di-(C2-C12)-alkylamino, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, (C1-C6)-alkoxycarbonyl, (C1-C6)-alkylcarbonyl, cyano, trifluoromethyl, trifluoromethyloxy, (C1-C6)-alkylsulfonyl, aminosulfonyl;
    • R2 is hydrogen, (C1-C6)-alkyl; or
    • R1 and R2 form together with the nitrogen atom bearing them a monocyclic, saturated or partially unsaturated 4- to 7-membered ring system or a bicyclic saturated or partially unsaturated 8- to 14-membered ring system, of which individual members of the ring systems may be replaced by one to three atoms or atomic groups from the series —CHR4-, —CR4R5-, —(C═R4)-, —NR6-, —C(═O)—, —O—, —S—, —SO—, —SO2—, with the proviso that two units from the series —O—, —S—, —SO—, —SO2— may not be adjacent;
    • R3 hydrogen, (C1-C6)-alkyl, benzyl;
    • R4, R5 identically or differently (C1-C6)-alkyl, halogen, trifluoromethyl, COOR3, cyclopropyl, cyclopropylene;
    • R6, R7 identically or differently hydrogen, (C1-C6)-alkyl, —(C6-C10)-aryl, (C5-C12)-heteroaryl, (C3-C12)-cycloalkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle;
      the tautomeric forms of the compounds and the physiologically tolerated salts thereof.
  • Preferred compounds of the formula I are those in which
    • R1 is (C5-C16)-alkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C3-C12)-cycloalkyl, (C8-C14)-bicycle, where aryl, heteroaryl, cycloalkyl or bicycle may be substituted one or more times by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, (C1-C6)-alkylmercapto, amino, (C1-C6)-alkylamino, di-(C2-C12-alkylamino, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, (C1-C6)-alkoxycarbonyl, (C1-C6)-alkylcarbonyl, cyano, trifluoromethyl, trifluoromethyloxy, (C1-C6)-alkylsulfonyl, aminosulfonyl.
  • Particularly preferred compounds of the formula I are those in which
    • X is identically or differently ═C(—R)— or ═N—, where one ═C(—R—)— is replaced by ═N—;
    • Y is —O—, —S—;
    • R is identically or differently hydrogen, halogen, (C1-C6)-alkyl, hydroxy, trifluoromethyl, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, COOR3, (C1-C6)-alkylsulfonyl, aminosulfonyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, (C1-C6)-alkylcarbonyl, CO—NR6R7, O—CO—NR6R7, O—CO—(C1-C6)-alkylene-CO—O—(C1-C6)-alkyl, O—CO—(C1-C6)-alkylene-CO—NR6R7 or unsubstituted or mono- or poly-F-substituted (C1-C6)-alkyloxy;
    • R1 is (C6-C12)-alkyl, (C1-C3)-alkylene-(C6-C10)-aryl, (C1-C3)-alkylene-(C5-C12)-heteroaryl, (C1-C3)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle, where aryl, heteroaryl, cycloalkyl or bicycle may be substituted one or more times by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, amino, (C1-C6)-alkylamino, trifluoromethyl;
    • R2 is hydrogen, (C1-C6)-alkyl; or
    • R1 and R2 together with the nitrogen atom bearing them are a monocyclic, saturated 5- to 6-membered ring system or a bicyclic saturated or partially unsaturated 9- to 10 membered ring system, of which individual members of the ring systems may be replaced by one to three atoms or atomic groups from the series —CHR4-, —CR4R5-, —(C═R4)-, —NR6-, —O—, —S—, with the proviso that two units from the series —O—, —S— may not be adjacent;
    • R3 is hydrogen, (C1-C6)-alkyl, benzyl;
    • R4, R5 are identically or differently (C1-C6)-alkyl, halogen, trifluoromethyl, COOR3, cyclopropyl, cyclopropylene;
    • R6, R7 are identically or differently hydrogen, (C1-C6)-alkyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, (C3-C12)-cycloalkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle.
  • Particularly preferred compounds of the formula I are also those in which
    • X is identically or differently ═C(—R)— or ═N—, where at least one ═C(—R—)— is replaced by ═N;
    • Y is —O— or —S—;
    • R is identically or differently hydrogen, halogen, (C1-C6)-alkyl, hydroxy, trifluoromethyl, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, COOR3, (C1-C6)-alkylsulfonyl, aminosulfonyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, (C1-C6)-alkylcarbonyl, CO—NR6R7, O—CO—NR6R7, O—CO—(C1-C6)-alkylene-CO—O—(C1-C6)-alkyl, O—CO—(C1-C6)-alkylene-CO—NR6R7 or unsubstituted or mono- or poly-F-substituted (C1-C6)-alkyloxy;
    • R1 is (C6-C12)-alkyl, (C1-C2)-alkylene-(C6-C10)-aryl, (C1-C2)-alkylene-(C5-C12)-heteroaryl, (C1-C2)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle, where aryl, heteroaryl, cycloalkyl or bicycle may be substituted one or more times by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, amino, (C1-C6)-alkylamino, trifluoromethyl;
    • R2 is hydrogen; or
    • R1 and R2 together with the nitrogen atom bearing them are a monocyclic, saturated 5- to 6-membered ring system or a bicyclic saturated or partially unsaturated 9- to 10 membered ring system, of which individual members of the ring systems may be replaced by one to three atoms or atomic groups from the series —CHR4-, —CR4R5-, —(C═R4)-, —NR6-, —O—, —S—, with the proviso that two units from the series —O—, —S— may not be adjacent;
    • R3 is hydrogen, (C1-C6)-alkyl, benzyl;
    • R4, R5 are identically or differently (C1-C6)-alkyl, halogen, trifluoromethyl, COOR3, cyclopropyl, cyclopropylene;
    • R6, R7 are identically or differently hydrogen, (C1-C6)-alkyl, —(C6-C10)-aryl, (C3-C12)-cycloalkyl, (C1-C4)-alkylene-(C6-C10)-aryl.
  • In a further particularly preferred embodiment of the compounds of the formula I,
    • R6, R7 are identically or differently hydrogen, (C1-C6)-alkyl or (C3-C6)-cycloalkyl.
  • Preferred compounds of the formula I are also those in which
    • X in position 5, 6 and 7 is identically or differently ═C(—R)—, in position 4 is ═N—.
  • Further preferred compounds of the formula I are those in which
    • X where appropriate in position 4 or 7 is ═C(—R)— with R=hydrogen, in position 5 or 6 is ═C(—R)— with R not hydrogen.
  • Very particularly preferred compounds of the formula I are those in which
    • Y is —O—, —S—;
    • R1 is (C6-C12)-alkyl, benzyl, (C1-C3)-alkylene-(C5-C12)-heteroaryl, (C1-C3)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle, where benzyl, heteroaryl, cycloalkyl or bicycle may be substituted by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy or trifluoromethyl;
    • R2 is hydrogen; or
    • R1 and R2 together with the nitrogen atom bearing them form a monocyclic, saturated 5- to 6-membered ring system, of which individual members of the ring systems may be replaced by one to two atoms or atomic groups from the series —CHR4-, —NR6-, in which R4 is as defined above, and R6 is (C1-C6)-alkyl or cyclopropyl.
  • In one embodiment of the compounds of the formula I,
    • X is identically or differently ═C(—R)— or ═N—, where one ═C(—R—)— is replaced by ═N—;
  • In one embodiment of the compounds of the formula I,
    • Y is —O—.
  • In another embodiment of the compounds of the formula I,
    • Y is —S—.
  • In a particular embodiment of the compounds of the formula I,
    • R2 is hydrogen.
  • Further very particularly preferred compounds of the formula I are those in which
    • Y is —O—;
    • R is identically or differently hydrogen, halogen, hydroxy or (C1-C6)-alkyl;
    • R1 is (C7-C12)-alkyl, benzyl, (C1-C3)-alkylene-(C5-C12)-heteroaryl, (C1-C3)-alkylene-(C4-C12)-cycloalkyl, (C8-C14)-bicycle, where benzyl, heteroaryl, cycloalkyl or bicycle may be substituted by halogen, (C1-C6)-alkyl, (C1-C3)-alkoxy or trifluoromethyl;
    • R2 is hydrogen.
  • Further very particularly preferred compounds of the formula I are also those in which
    • Y is —O—;
    • R is identically or differently hydrogen, halogen or (C1-C6)-alkyl;
    • R1 is (C6-C12)-alkyl, benzyl, —CH2—(C5-C12)-heteroaryl, —CH2—(C4-C12)-cycloalkyl, (C8-C14)-bicycle, where benzyl, heteroaryl, cycloalkyl or bicycle may be substituted by halogen, (C1-C6)-alkyl, (C1-C3)-alkoxy or trifluoromethyl;
    • R2 is hydrogen.
  • Further very particularly preferred compounds of the formula I are also those in which
    • Y is —O—;
    • R is identically or differently hydrogen, halogen or (C1-C6)-alkyl;
    • R1 is (C6-C12)-alkyl, benzyl, —CH2—(C4-C12)-cyclohexyl, indanyl, tetrahydronaphthyl, where benzyl, cyclohexyl, indanyl or tetrahydronaphthyl may be substituted by (C1-C6)-alkyl;
    • R2 is hydrogen.
  • The invention relates to compounds of the formula I in the form of their salts, racemates, racemic mixtures and pure enantiomers, and to their diastereomers and mixtures thereof.
  • The alkyl and alkylene radicals in the substituents R, R1, R2, R3, R4, R5, R6 and R7 may be either straight-chain or branched. Halogen is fluorine, chlorine, bromine or iodine, in particular fluorine or chlorine. The alkyl and alkylene radicals may be substituted one or more times by suitable groups
  • Aryl means an aromatic carbocyclic mono- or bicyclic ring system which comprises 6 to 10 atoms in the ring or in the rings.
  • The aryl radicals may be substituted one or more times by suitable groups.
  • Heteroaryl is a mono- or bicyclic aromatic ring system having 5 to 12 ring members, in which at least one atom in the ring system is a heteroatom from the series N, O and S.
  • Suitable “heteroaryl rings” or “heteroaryl radicals” are, for example, benzimidazolyl, benzofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, quinolinyl, furyl, furazanyl, imidazolyl, 1H-indazolyl, indolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyridyl, pyrrolyl, thiazolyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thiophenyl.
  • Preferred heteroaryl radicals are thiophenyl, pyrazolyl and pyridyl. The heteroaryl rings or heteroaryl radicals may be substituted one or more times by suitable groups.
  • A cycloalkyl radical is a ring system which comprises one or more rings and which is in saturated or partially unsaturated (with one or two double bonds) form and which is composed exclusively of carbon atoms.
  • The cycloalkyl radicals may be substituted one or more times by suitable groups.
  • Bicycle means bicyclic ring systems which are in saturated or partially unsaturated form and which, apart from carbon, may also comprise one or more heteroatoms such as, for example, nitrogen, oxygen or sulfur. This definition also includes ring systems which comprise a fused benzene nucleus. Examples which may be mentioned are the tetrahydronaphthyl, alpha- or beta-tetralon-, indanyl- or indan-1-on-yl radical. Preferred bicyclic radicals are those of formula Ic
  • Figure US20080167339A1-20080710-C00003
  • with q=1 or 2. The bicyclic radicals may be substituted one or more times by suitable groups.
  • Pharmaceutically acceptable salts are, because their solubility in water is greater than that of the initial or basic compounds, particularly suitable for medical applications. These salts must have a pharmaceutically acceptable anion or cation. Suitable pharmaceutically acceptable acid addition salts of the compounds of the invention are salts of inorganic acids such as hydrochloric acid, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acid, and of organic acids such as, for example, acetic acid, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isethionic, lactic, lactobionic, maleic, malic, methanesulfonic, succinic, p-toluenesulfonic and tartaric acid. Suitable pharmaceutically acceptable basic salts are ammonium salts, alkali metal salts (such as sodium and potassium salts) and alkaline earth metal salts (such as magnesium and calcium salts) and salts of trometamol (2-amino-2-hydroxymethyl-1,3-propanediol), diethanolamine, lysine or ethylenediamine.
  • Salts with a pharmaceutically unacceptable anion such as, for example, trifluoroacetate likewise belong within the framework of the invention as useful intermediates for the preparation or purification of pharmaceutically acceptable salts and/or for use in nontherapeutic, for example in vitro, applications.
  • The term “physiologically functional derivative” used herein refers to any physiologically tolerated derivative of a compound of the invention of the formula I, for example an ester, which on administration to a mammal such as, for example, a human is able to form (directly or indirectly) a compound of the formula I or an active metabolite thereof.
  • Physiologically functional derivatives also include prodrugs of the compounds of the invention as, for example, described in H. Okada et al., Chem. Pharm. Bull. 1994, 42, 57-61. Such prodrugs can be metabolized in vivo to a compound of the invention. These prodrugs may themselves be active or not.
  • The compounds of the invention may also exist in various polymorphous forms, for example as amorphous and crystalline polymorphous forms. All polymorphous forms of the compounds of the invention belong within the framework of the invention and are a further aspect of the invention.
  • All references to “compound(s) of formula I” hereinafter refer to compound(s) of the formula I as described above, and their salts, solvates and physiologically functional derivatives as described herein.
  • Use
  • The compounds of the invention of the general formula I have a surprising inhibitory effect on hormone sensitive lipase, HSL, an allosteric enzyme in adipocytes which is inhibited by insulin and is responsible for the breakdown of fats in fat cells and thus for transferring fat constituents into the blood stream. Inhibition of this enzyme is therefore equivalent to an insulin-like effect of the compounds of the invention, eventually leading to a reduction of free fatty acids in the blood and of blood glucose. They can therefore be employed for metabolic derangements such as, for example, for non-insulin-dependent diabetes mellitus, for diabetic syndrome and for direct pancreatic damage.
  • The compounds of the invention of the general formula I, especially those in which R2 is hydrogen, may additionally have an inhibitory effect on endothelial lipase (EL). The preferred substrate for EL is HDL, which has antiatherosclerotic activity. A reduction in the HDL level leads to progression of atherosclerosis and its sequelae such as coronary heart disease and moreover favors development of the metabolic syndrome and its sequelae diabetes. An inhibition of EL should thus generally lead to prevention of atherosclerotic disorders and indirectly reduce the probability of people with an increased risk for diabetes becoming ill.
  • It has further been found that the inhibitory effect of the compounds of the invention of the general formula I is selective in relation to other lipases.
  • The compounds of the invention of the formula I may also have an inhibitory effect on triglyceride lipase.
  • Compounds of this type are particularly suitable for the treatment and/or prevention of
    • 1.—Disorders of fatty acid metabolism and glucose utilization disorders
    • 2.—Disorders of the insulin sensitivity of myo-, adipo- and hepatocytes (insulin resistance)-metabolic syndrome
    • 3. Diabetes mellitus, especially type 2 diabetes, including the prevention of the sequelae associated therewith.
    •  Particular aspects in this connection are
      • hyperglycemia,
      • improvement in insulin resistance,
      • improvement in glucose tolerance,
      • protection of the pancreatic β cells
      • prevention of macro- and microvascular disorders
    • 4. Dyslipidemias and the sequelae thereof such as, for example, atherosclerosis, coronary heart disease, cerebrovascular disorders etc., especially those (but not restricted thereto) which are characterized by one or more of the following factors:
      • high plasma triglyceride concentrations, high postprandial plasma triglyceride concentrations
      • low HDL cholesterol concentration
      • low apoA lipoprotein concentrations
      • high LDL cholesterol concentrations
      • small dense LDL cholesterol particles
      • high apoB lipoprotein concentrations
    • 5. Various other conditions which may be associated with the metabolic syndrome, such as:
      • obesity (excess weight), including central obesity
      • thromboses, hypercoagulable and prothrombotic stages (arterial and venous)
      • high blood pressure
      • heart failure such as, for example (but not restricted thereto), following myocardial infarction, hypertensive heart disease or cardiomyopathy
    • 6. Other disorders or conditions in which inflammatory reactions or cell differentiation may for example be involved are:
      • atherosclerosis such as, for example (but not restricted thereto), coronary sclerosis including angina pectoris or myocardial infarction, stroke
      • vascular restenosis or reocclusion
      • chronic inflammatory bowel diseases such as, for example, Crohn's disease and ulcerative colitis
      • pancreatitis
      • other inflammatory states
      • retinopathy
      • adipose cell tumors
      • adipose cell carcinomas such as, for example, liposarcoma
      • solid tumors and neoplasms such as, for example (but not restricted thereto), carcinomas of the gastrointestinal tract, of the liver, of the biliary tract and of the pancreas, endocrine tumors, carcinomas of the lungs, of the kidneys and of the urinary tract, of the genital tract, prostate carcinomas etc.
      • acute and chronic myeloproliferative disorders and lymphomas
      • angiogenesis
      • neurodegenerative disorders
      • Alzheimer's disease
      • multiple sclerosis
      • Parkinson's disease
      • erythemato-squamous dermatoses such as, for example, psoriasis
      • acne vulgaris
      • other skin disorders and dermatological conditions which are modulated by PPAR
      • eczemas and neurodermatitis
      • dermatitis such as, for example, seborrheic dermatitis or photodermatitis
      • keratitis and keratosis such as, for example, seborrheic keratoses, senile keratoses, actinic keratosis, photo-induced keratoses or keratosis follicularis
      • keloids and keloid prophylaxis
      • warts, including condylomata or condylomata acuminata
      • human papilloma viral (HPV) infections, such as, for example, venereal papillomata, viral warts such as, for example, molluscum contagiosum, leukoplakia
      • papular dermatoses such as, for example, lichen planus
      • skin cancer such as, for example, basal cell carcinoma, melanomas or cutaneous T-cell lymphomas
      • localized benign epidermal tumors such as, for example, keratoderma, epidermal naevi
      • chilblains
      • high blood pressure
      • syndrome X
      • polycystic ovary syndrome (PCOS)
      • asthma
      • osteoarthritis
      • lupus erythematosus (LE) or inflammatory rheumatic disorders such as, for example, rheumatoid arthritis
      • vasculitis
      • wasting (cachexia)
      • gout
      • ischemia/reperfusion syndrome
      • acute respiratory distress syndrome (ARDS)
    Formulations
  • The amount of a compound of the invention necessary to achieve the desired biological effect depends on a number of factors, for example the specific compound chosen, the intended use, the mode of administration and the clinical condition of the patient. The daily dose is generally in the range from 0.3 mg to 100 mg (typically from 3 mg to 50 mg) per day and per kilogram of body weight, for example 3-10 mg/kg/day. An intravenous dose may be, for example, in the range from 0.3 mg to 1.0 mg/kg, which can suitably be administered as infusion of 10 ng to 100 ng per kilogram and per minute. Suitable infusion solutions for these purposes may contain, for example, from 0.1 ng to 10 mg, typically from 1 ng to 10 mg, per milliliter. Single doses may contain, for example, from 1 mg to 10 g of the active ingredient. Thus, ampoules for injections may contain, for example, from 1 mg to 100 mg, and single-dose formulations which can be administered orally, such as, for example, tablets or capsules, may contain, for example, from 0.05 to 1000 mg, typically from 0.5 to 600 mg. For the therapy of the abovementioned conditions, the compounds of formula I may be used as the compound itself, but they are preferably in the form of a pharmaceutical composition with an acceptable carrier. The carrier must, of course, be acceptable in the sense that it is compatible with the other ingredients of the composition and is not harmful for the patient's health. The carrier may be a solid or a liquid or both and is preferably formulated with the compound as a single dose, for example as a tablet, which may contain from 0.05% to 95% by weight of the active ingredient. Other pharmaceutically active substances may likewise be present, including other compounds of the invention. The pharmaceutical compositions of the invention can be produced by one of the known pharmaceutical methods, which essentially consist of mixing the ingredients with pharmacologically acceptable carriers and/or excipients.
  • Pharmaceutical compositions of the invention are those suitable for oral, rectal, topical, peroral (for example sublingual) and parenteral (for example subcutaneous, intramuscular, intradermal or intravenous) administration, although the most suitable mode of administration depends in each individual case on the nature and severity of the condition to be treated and on the nature of the compound of formula I used in each case. Coated formulations and coated slow-release formulations also belong within the framework of the invention. Preference is given to acid- and gastric juice-resistant formulations. Suitable coatings resistant to gastric juice comprise cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropylmethylcellulose phthalate and anionic polymers of methacrylic acid and methyl methacrylate.
  • Suitable pharmaceutical preparations for oral administration may be in the form of separate units such as, for example, capsules, cachets, suckable tablets or tablets, each of which contain a defined amount of the compound of formula I; as powders or granules; as solution or suspension in an aqueous or nonaqueous liquid; or as an oil-in-water or water-in-oil emulsion. These compositions may, as already mentioned, be prepared by any suitable pharmaceutical method which includes a step in which the active ingredient and the carrier (which may consist of one or more additional ingredients) are brought into contact. The compositions are generally produced by uniform and homogeneous mixing of the active ingredient with a liquid and/or finely divided solid carrier, after which the product is shaped if necessary. Thus, for example, a tablet can be produced by compressing or molding a powder or granules of the compound, where appropriate with one or more additional ingredients. Compressed tablets can be produced by tableting the compound in free-flowing form such as, for example, a powder or granules, where appropriate mixed with a binder, glidant, inert diluent and/or one or more surface-active/dispersing agent(s) in a suitable machine. Molded tablets can be produced by molding the compound, which is in powder form and is moistened with an inert liquid diluent, in a suitable machine.
  • Pharmaceutical compositions which are suitable for peroral (sublingual) administration comprise suckable tablets which contain a compound of formula I with a flavoring, normally sucrose and gum arabic or tragacanth, and pastilles which comprise the compound in an inert base such as gelatin and glycerol or sucrose and gum arabic.
  • Pharmaceutical compositions suitable for parenteral administration comprise preferably sterile aqueous preparations of a compound of formula I, which are preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although administration may also take place by subcutaneous, intramuscular or intradermal injection. These preparations can preferably be produced by mixing the compound with water and making the resulting solution sterile and isotonic with blood. Injectable compositions of the invention generally contain from 0.1 to 5% by weight of the active compound.
  • Pharmaceutical compositions suitable for rectal administration are preferably in the form of single-dose suppositories. These can be produced by mixing a compound of the formula I with one or more conventional solid carriers, for example cocoa butter, and shaping the resulting mixture.
  • Pharmaceutical compositions suitable for topical use on the skin are preferably in the form of ointment, cream, lotion, paste, spray, aerosol or oil. Carriers which can be used are petrolatum, lanolin, polyethylene glycols, alcohols and combinations of two or more of these substances. The active ingredient is generally present in a concentration of from 0.1 to 15% by weight of the composition, for example from 0.5 to 2%.
  • Transdermal administration is also possible. Pharmaceutical compositions suitable for transdermal uses can be in the form of single patches which are suitable for long-term close contact with the patient's epidermis. Such patches suitably contain the active ingredient in an aqueous solution which is buffered where appropriate, dissolved and/or dispersed in an adhesive or dispersed in a polymer. A suitable active ingredient concentration is about 1% to 35%, preferably about 3% to 15%. A particular possibility is for the active ingredient to be released by electrotransport or iontophoresis as described, for example, in Pharmaceutical Research, 2(6): 318 (1986).
  • The compounds of the formula I with R2 hydrogen are distinguished by favorable effects on disorders of lipid metabolism. They beneficially influence the HDL to LDL ratio and increase in particular the HDL level and are suitable for the prevention and treatment of dyslipidemias and metabolic syndrome and their diverse sequelae such as atherosclerosis, coronary heart disease, heart failure, obesity and diabetes.
  • The compounds of the formula I with R1 and R2, which form a ring system, are distinguished by favorable effects on metabolic disorders. They beneficially influence lipid and sugar metabolism, in particular they lower the triglyceride level and are suitable for the prevention and treatment of type II diabetes and arteriosclerosis and the diverse sequelae thereof.
  • Combinations with Other Medicaments
  • The compounds of the invention can be administered alone or in combination with one or more further pharmacologically active substances which have, for example, favorable effects on metabolic disturbances or disorders frequently associated therewith. Examples of such medicaments are
      • 1. medicaments which lower blood glucose, antidiabetics,
      • 2. active ingredients for the treatment of dyslipidemias,
      • 3. antiatherosclerotic medicaments,
      • 4. antiobesity agents,
      • 5. antiinflammatory active ingredients
      • 6. active ingredients for the treatment of malignant tumors
      • 7. antithrombotic active ingredients
      • 8. active ingredients for the treatment of high blood pressure
      • 9. active ingredients for the treatment of heart failure and
      • 10. active ingredients for the treatment and/or prevention of complications caused by diabetes or associated with diabetes.
  • They can be combined with the compounds of the invention of the formula I in particular for a synergistic improvement in the effect. Administration of the active ingredient combination can take place either by separate administration of the active ingredients to the patient or in the form of combination products in which a plurality of active ingredients are present in one pharmaceutical preparation.
  • Examples which may be mentioned are:
  • Antidiabetics
  • Suitable antidiabetics are disclosed for example in the Rote Liste 2001, chapter 12 or in the USP Dictionary of USAN and International Drug Names, US Pharmacopeia, Rockville 2003. Antidiabetics include all insulins and insulin derivatives such as, for example, Lantus® (see www.lantus.com) or Apidra®, and other fast-acting insulins (see U.S. Pat. No. 6,221,633), GLP-1 receptor modulators as described in WO 01/04146 or else, for example, those disclosed in WO 98/08871 of Novo Nordisk A/S. The orally effective hypoglycemic active ingredients include, preferably, the sulfonylureas which act on the ATP-dependent potassium channel of the beta cells (e.g. disclosed in WO 97/26265 and WO 99/03861), biguanides, meglitinides, glucagon antagonists, oral GLP-1 agonists, DPP-IV inhibitors, insulin sensitizers, e.g. PPAR and PXR modulators and active ingredients such as, for example, oxadiazolidinediones, thiazolidinediones, inhibitors of liver enzymes which are involved in stimulating gluconeogenesis and/or glycogenolysis, modulators of glucose uptake such as, for example, glucosidase inhibitors, compounds which alter lipid metabolism and lead to a change in the blood lipid composition, compounds which reduce food intake or food uptake.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with insulin.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with substances which influence hepatic glucose production such as, for example, glycogen phosphorylase inhibitors (see: WO 01/94300, WO 02/096864, WO 03/084923, WO 03/084922, WO 03/104188).
  • In one embodiment, the compounds of the formula I are administered in combination with an active ingredient which acts on the ATP-dependent potassium channel of the beta cells, such as, for example, sulfonylureas (e.g. tolbutamide, glibenclamide, glipizide, glimepiride) or glinides (e.g. repaglinide).
  • In one embodiment, the compounds of the formula I are administered in combination with a biguanide such as, for example, metformin.
  • In one embodiment, the compounds of the formula I are administered in combination with a PPARgamma agonist or thiazolidinedione such as, for example, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed in WO 97/41097 of Dr. Reddy's Research Foundation, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2-quinazolinylmethoxy]phenyl]methyl]-2,4-thiazolidinedione.
  • In one embodiment, the compounds of the formula I are administered in combination with a DPPIV inhibitor as described, for example, in WO98/19998, WO99/61431, WO99/67278, WO99/67279, WO01/72290, WO 02/38541, WO03/040174, in particular P 93/01 (1-cyclopentyl-3-methyl-1-oxo-2-pentanammonium chloride), P-31/98, LAF237 (1-[2-[3-hydroxyadamant-1-ylamino)acetyl]pyrrolidine-2-(S)-carbonitrile), TS021 ((2S,4S)-4-fluoro-1-[[(2-hydroxy-1,1-dimethylethyl)amino]-acetyl]pyrrolidine-2-carbonitrile monobenzenesulfonate).
  • In one embodiment, the compounds of the formula I are administered in combination with compounds with an inhibitory effect on SGLT-1 and/or 2, as disclosed directly or indirectly for example in WO2004/007517, WO2004/052902, WO2004/052903 and WO2005/121161.
  • In one embodiment, the compounds of the formula I are administered in combination with an α-glucosidase inhibitor such as, for example, miglitol or acarbose.
  • In one embodiment, the compounds of the formula I are administered in combination with more than one of the aforementioned compounds, e.g. in combination with a sulfonylurea and metformin, a sulfonylurea and acarbose, repaglinide and metformin, insulin and a sulfonylurea, insulin and metformin, insulin and troglitazone, insulin and lovastatin, etc.
  • Lipid Modulators
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an HMGCoA reductase inhibitor such as lovastatin, fluvastatin, pravastatin, simvastatin, ivastatin, itavastatin, atorvastatin, rosuvastatin.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a bile acid absorption inhibitor (see, for example, U.S. Pat. No. 6,245,744, U.S. Pat. No. 6,221,897, U.S. Pat. No. 6,277,831, EP 0683 773, EP 0683 774).
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a polymeric bile acid adsorbent such as, for example, cholestyramine, colesevelam.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a cholesterol resorption inhibitor as described for example in WO 0250027, or ezetimibe, tiqueside, pamaqueside.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an LDL receptor inducer (see, for example, U.S. Pat. No. 6,342,512).
  • In one embodiment, the compounds of the formula I are administered in combination with bulking agents, preferably insoluble bulking agents (see, for example, carob/Caromax® (Zunft H J; et al., Carob pulp preparation for treatment of hypercholesterolemia, ADVANCES IN THERAPY (2001 September-October), 18(5), 230-6)). Caromax is a carob-containing product from Nutrinova, Nutrition Specialties & Food Ingredients GmbH, Industriepark Hoechst, 65926 Frankfurt/Main). Combination with Caromax® is possible in one preparation or by separate administration of compounds of the formula I and Caromax®. Caromax® can in this connection also be administered in the form of food products such as, for example, in bakery products or muesli bars.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a PPARalpha agonist.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a mixed PPAR alpha/gamma agonist such as, for example, AZ 242 (tesaglitazar, (S)-3-(4-[2-(4-methanesulfonyloxyphenyl)ethoxy]-phenyl)-2-ethoxypropionic acid), BMS 298585 (N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine) or as described in WO 99/62872, WO 99/62871, WO 01/40171, WO 01/40169, WO96/38428, WO 01/81327, WO 01/21602, WO 03/020269, WO 00/64888 or WO 00/64876.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a fibrate such as, for example, fenofibrate, gemfibrozil, clofibrate, bezafibrate.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with nicotinic acid or niacin.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a CETP inhibitor, e.g. CP-529, 414 (torcetrapib).
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an ACAT inhibitor.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an MTP inhibitor such as, for example, implitapide.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an antioxidant.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a lipoprotein lipase inhibitor.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with an ATP citrate lyase inhibitor.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a squalene synthetase inhibitor.
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a lipoprotein(a) antagonist.
  • Antiobesity Agents
  • In one embodiment of the invention, the compounds of the formula I are administered in combination with a lipase inhibitor such as, for example, orlistat.
  • In one embodiment, the further active ingredient is fenfluramine or dexfenfluramine. In another embodiment, the further active ingredient is sibutramine.
  • In a further embodiment, the compounds of the formula I are administered in combination with CART modulators (see “Cocaine-amphetamine-regulated transcript influences energy metabolism, anxiety and gastric emptying in mice” Asakawa, A, et al., M.: Hormone and Metabolic Research (2001), 33(9), 554-558), NPY antagonists, e.g. naphthalene-1-sulfonic acid {4-[(4-aminoquinazolin-2-ylamino)methyl]cyclohexylmethyl}amide; hydrochloride (CG P 71683A)), MC4 agonists (e.g. 1-amino-1,2,3,4-tetrahydronaphthalene-2-carboxylic acid [2-(3a-benzyl-2-methyl-3-oxo-2,3,3a,4,6,7-hexahydropyrazolo[4,3-c]pyridin-5-yl)-1-(4-chlorophenyl)-2-oxoethyl]amide; (WO 01/91752)), orexin antagonists (e.g. 1-(2-methylbenzoxazol-6-yl)-3-[1,5]naphthyridin-4-ylurea; hydrochloride (SB-334867-A)), H3 agonists (3-cyclohexyl-1-(4,4-dimethyl-1,4,6,7-tetrahydroimidazo[4,5-c]pyridin-5-yl)propan-1-one oxalic acid salt (WO 00/63208)); TNF agonists, CRF antagonists (e.g. [2-methyl-9-(2,4,6-trimethylphenyl)-9H-1,3,9-triazafluoren-4-yl]dipropylamine (WO 00/66585)), CRF BP antagonists (e.g. urocortin), urocortin agonists, β3 agonists (e.g. 1-(4-chloro-3-methanesulfonylmethylphenyl)-2-[2-(2,3-dimethyl-1H-indol-6-yloxy)ethylamino]ethanol; hydrochloride (WO 01/83451)), MSH (melanocyte-stimulating hormone) agonists, CCK-A agonists (e.g. {2-[4-(4-chloro-2,5-dimethoxyphenyl)-5-(2-cyclohexylethyl)thiazol-2-ylcarbamoyl]-5,7-dimethylindol-1-yl}acetic acid trifluoroacetic acid salt (WO 99/15525)), serotonin reuptake inhibitors (e.g. dexfenfluramine), mixed serotoninergic and noradrenergic compounds (e.g. WO 00/71549), 5HT agonists e.g. 1-(3-ethylbenzofuran-7-yl)piperazine oxalic acid salt (WO 01/09111), bombesin agonists, galanin antagonists, growth hormone (e.g. human growth hormone), growth hormone-releasing compounds (6-benzyloxy-1-(2-diisopropylaminoethylcarbamoyl)-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tertiary butyl ester (WO 01/85695)), TRH agonists (see, for example, EP 0 462 884), uncoupling protein 2 or 3 modulators, leptin agonists (see, for example, Lee, Daniel W.; Leinung, Matthew C.; Rozhavskaya-Arena, Marina; Grasso, Patricia. Leptin agonists as a potential approach to the treatment of obesity. Drugs of the Future (2001), 26(9), 873-881), DA agonists (bromocriptine, Doprexin), lipase/amylase inhibitors (e.g. WO 00/40569), PPAR modulators (e.g. WO 00/78312), RXR modulators or TR-β agonists.
  • In another embodiment, the further active ingredient is a cannabinoid receptor 1 antagonist (such as, for example, rimonabant, SR147778 or those as are described in, for example, EP 0656354, WO 00/15609, WO 02/076949, WO2005080345, WO2005080328, WO2005/080343, WO2005/075450, WO2005/080357, WO2001/70700, WO2003/026647-48, WO2003/02776, WO2003/040107, WO2003/007887, WO2003/027069, U.S. Pat. No. 6,509,367, WO2001/32663, WO2003/086288, WO2003/087037, WO2004/048317, WO2004/058145, WO2003/084930, WO2003/084943, WO2004/058744, WO2004/013120, WO2004/029204, WO2004/035566, WO2004/058249, WO2004/058255, WO2004/058727, WO2004/069838, US2004/0214837, US2004/0214855, US2004/0214856, WO2004/096209, WO2004/096763, WO2004/096794, WO2005/000809, WO2004/099157, US2004/0266845, WO2004/110453, WO2004/108728, WO2004/000817, WO2005/000820, US2005/0009870, WO2005/00974, WO2004/111033-34, WO2004/11038-39, WO2005/016286, WO2005/007111, WO2005/007628, US2005/0054679, WO2005/027837, WO2005/028456, WO2005/063761-62, WO2005/061509, WO2005/077897).
  • In one embodiment of the invention, the further active ingredient is leptin.
  • In one embodiment, the further active ingredient is dexamphetamine, amphetamine, mazindole or phentermine.
  • In one embodiment, the compounds of the formula I are administered in combination with medicaments having effects on the coronary circulation and the vascular system, such as, for example, ACE inhibitors (e.g. ramipril), medicaments which act on the angiotensin-renine system, calcium antagonists, beta blockers etc.
  • In one embodiment, the compounds of the formula I are administered in combination with medicaments having an antiinflammatory effect.
  • In one embodiment, the compounds of the formula I are administered in combination with medicaments which are employed for cancer therapy and cancer prevention.
  • It will be appreciated that every suitable combination of the compounds of the invention with one or more of the aforementioned compounds and optionally one or more other pharmacologically active substances is regarded as falling within the protection conferred by the present invention.
  • The activity of the compounds of the invention of the formula I was tested in the following enzyme assay systems:
  • 1. HSL Inhibition Assay 1.1. Preparation of the Partially Purified HSL:
  • Isolated rat fat cells are obtained from epididymal adipose tissue from untreated male rats (Wistar, 220-250 g) by collagenase treatment in accordance with published methods (e.g. S, Nilsson et al., Anal. Biochem. 158, 1986, 399-407; G. Fredrikson et al., J. Biol. Chem. 256, 1981, 6311-6320; H. Tornquist et al., J. Biol. Chem. 251, 1976, 813-819). The fat cells from 10 rats are washed three times by flotation with 50 ml of homogenization buffer (25 ml Tris/HCl, pH 7.4, 0.25 M sucrose, 1 mM ETDA, 1 mM DTT, 10 μg/ml leupeptin, 10 μg/ml antipain, 20 μg/ml pepstatin) each time and finally taken up in 10 ml of homogenization buffer. The fat cells are homogenized in a Teflon-in-glass homogenizer (Braun-Melsungen) by 10 strokes at 1500 rpm and 15° C. The homogenate is centrifuged (Sorvall SM24 tubes, 5000 rpm, 10 min, 4° C.). The subnatant between the layer of fat at the top and the pellet is removed and the centrifugation is repeated. The subnatant resulting therefrom is centrifuged again (Sorvall SM24 tubes, 20 000 rpm, 45 min, 4° C.). The subnatant is removed, and 1 g of heparin-Sepharose (Pharmacia-Biotech, CL-6B, washed 5× with 25 mM Tris/HCl, pH 7.4, 150 mM NaCl) is added. After incubation at 4° C. for 60 min (shaking at intervals of 15 min), the mixture is centrifuged (Sorvall SM24 tubes, 3000 rpm, 10 min, 4° C.). The supernatant is adjusted to pH 5.2 by adding glacial acetic acid and is incubated at 4° C. for 30 min. The precipitates are collected by centrifugation (Sorvall SS34, 12 000 rpm, 10 min, 4° C.) and suspended in 2.5 ml of 20 mM Tris/HCl, pH 7.0, 1 mM EDTA, 65 mM NaCl, 13% sucrose, 1 mM DTT, 10 μg/ml leupeptin/pepstatin/antipain. The suspension is dialyzed against 25 mM Tris/HCl, pH 7.4, 50% glycerol, 1 mM DTT, 10 μg/ml leupeptin, pepstatin, antipain at 4° C. overnight and then loaded onto a hydroxiapatite column (0.1 g per 1 ml of suspension, equilibrated with 10 mM potassium phosphate, pH 7.0, 30% glycerol, 1 mM DTT). The column is washed with four volumes of equilibration buffer at a flow rate of 20 to 30 ml/h. The HSL is eluted with one volume of equilibration buffer containing 0.5 M potassium phosphate and then dialyzed (see above) and concentrated 5- to 10-fold by ultrafiltration (Amicon Diaflo PM 10 Filter) at 4° C. The partially purified HSL can be stored at −70° C. for 4 to 6 weeks.
  • 1.2 HSL Activity Assay:
  • To prepare the substrate, 25-50 μCi of [3H]trioleoylglycerol (in toluene), 6.8 μmol of unlabeled trioleoylglycerol and 0.6 mg of phospholipids (phosphatidylcholine/phosphatidylinositol 3:1 w/v) are mixed, dried with N2 and then taken up in 2 ml of 0.1 M KPi (pH 7.0) by ultrasound treatment (Branson 250, microtip, setting 1-2, 2×1 min with an interval of 1 min). After addition of 1 ml of KPi and renewed ultrasound treatment (4×30 sec on ice with intervals of 30 sec), 1 ml of 20% BSA (in KPi) is added (final concentration of trioleoylglycerol 1.7 mM). For the reaction, 100 μl of substrate solution are pipetted into 100 μl of HSL solution (HSL prepared as above, diluted in 20 mM KPi, pH 7.0, 1 mM EDTA, 1 mM DTT, 0.02% BSA, 20 μg/ml pepstatin, 10 μg/ml leupeptin) and incubated at 37° C. for 30 min. Addition of 3.25 ml of methanol/chloroform/heptane (10:9:7) and of 1.05 ml of 0.1 M K2CO3, 0.1 M boric acid (pH 10.5) is followed by thorough mixing and finally centrifugation (800×g, 20 min). After phase separation, one equivalent of the upper phase (1 ml) is removed and the radioactivity is determined by liquid scintillation measurement.
  • 1.3 Evaluation of the HSL-Inhibitory Effect:
  • Substances are normally tested in four independent mixtures. The inhibition of the HSL enzymatic activity by a test substance is determined by comparing with an uninhibited control reaction. The IC50 is calculated from an inhibition plot with at least 10 concentrations of the test substance. The GRAPHIT, Elsevier-BIOSOFT software package is used to analyze the data.
  • 2. EL Inhibition Assay: 2.1. Preparation of EL
  • EL is released as secretory protein in high concentration into cell culture medium (conditioned medium) by recombinant cell lines (CHO, HEK293). This is employed as enzyme solution after concentration.
  • 2.2. EL Activity Assay
  • The phospholipase-specific substrate 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phosphocholine, (manufacturer Molecular Probes) is used to characterize the enzymatic activity of endothelial lipase and the effect of inhibitors. Hydrolysis of the A1 ester linkage of this phospholipid by the enzyme liberates the fluorescent dye Bodipy which can be detected after separation by thin-layer chromatography on an HPTLC plate (silica gel 60, Merck) or directly in the reaction vessel by measuring the fluorescence.
  • The substrate solution is prepared by taking up 100 μg of 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phospho-choline (manufacturer Molecular Probes), 2.4 mg of tripalmitin (Sigma) and 7.9 mg of DOP-choline (1,2-dioleoyl-sn-glycero-3-phosphocholine) in 393 μl of chloroform and then transferring 157 μl into a fresh reaction vessel. After evaporation of the solvent, the lipid mixture is dissolved in 4 ml of 200 mM TRIS-HCl, 150 mM sodium chloride, pH=7.4, by sonication twice. The subsequent enzymic reaction takes place at 37° C. for 60 minutes. For this purpose, 45 μl of the substrate solution are incubated with 1 μl of inhibitor of appropriate concentration (dissolved in DMSO, pure DMSO solution is used as control) and 5 μl of enzyme solution (conditioned medium). Then 3 μl of the assay mixture are loaded onto an HPTLC plate (silica gel 60, Merck), and the liberated fluorescent dye is separated for detection with an eluent (diethyl ether:petroleum benzine:acetic acid [78:22:1]). After evaporation of the eluent, the plate is read in a fluorescence scanner. An increased liberation of the fluorescent dye in the uninhibited reaction is to be observed as a measure of the enzymic activity.
  • 2.3. Evaluation of the EL-Inhibitory Effect:
  • The enzymatic activity is reduced as a function of the inhibitor concentration used, and the inhibitor concentration at which a half-maximum enzymatic activity is observed is called IC50.
  • 2.4. Further EL Inhibition Assay:
  • The phospholipase-specific substrate 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phosphocholine, (manufacturer Molecular Probes) is used to characterize the enzymatic activity of endothelial lipase and the effect of inhibitors. Hydrolysis of the A1 ester linkage of this phospholipid by the enzyme liberates the fluorescent dye Bodipy which can be detected after separation by thin-layer chromatography on an HPTLC plate (silica gel 60, Merck) or directly in the reaction vessel by measuring the fluorescence. The substrate solution is prepared by dissolving 100 μg of 1,2-bis(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-undecanoyl)-sn-glycero-3-phospho-choline (manufacturer Molecular Probes) in 100 μl of DMSO and taking up in 2.4 mg of tripalmitin (Sigma) in 393 μl of chloroform which comprises 20 mg/ml DOP-choline (1,2-dioleoyl-sn-glycero-3-phosphocholine). 39.3 μl of this lipid mixture are transferred into a fresh reaction vessel, and the solvent is evaporated off. The lipid mixture is dissolved in 4 ml of 200 mM TRIS-HCl, 150 mM sodium chloride, pH=7.4, by sonication twice. The subsequent enzymic reaction takes place at 37° C. for 90 minutes. For this purpose, 20 μl of the substrate solution are incubated with 2 μl of inhibitor of appropriate concentration (dissolved in 10% DMSO, using 10% strength DMSO solution for control) and 2 μl of enzyme solution (conditioned medium). The 4 μl of the assay mixture are loaded onto an HPTLC plate (silica gel 60, Merck), and the liberated fluorescent dye is separated for detection with an eluent (diethyl ether:petroleum benzine:acetic acid [78:22:1]). After evaporation of the eluent, the plate is read in a fluorescence scanner. An increased liberation of the fluorescent dye in the uninhibited reaction is to be observed as a measure of the enzymic activity.
  • In this assay, the compounds of the examples showed the following IC50 values:
  • IC50 [μM]
    Example EL
    1 0.05
    2 0.01
    3 0.069
    4 0.39
    5 0.01
    9 0.011
    11 0.354
    13 0.0005
    14 0.18
    15 0.23
    16 0.18
    17 0.061
    18 0.48
    19 0.083
    20 0.09
  • Preparation Processes
  • The compounds of the invention of the general formula I are prepared by methods known per se, e.g. by acylation of substituted or unsubstituted azolopyridin-2-one derivatives II with carbamoyl chlorides III (method A), or in two stages by reacting azolopyridin-2-one derivatives II with phosgene or equivalents such as trichloromethyl chlorocarbonate, ditrichloromethyl carbonate or 4-nitrophenyl chloroformate and further reaction of the resulting azolopyridin-2-onecarboxylic acid derivative with amines IV (method B). For compounds in which R2 is hydrogen, the azolopyridin-2-one derivatives II can also be reacted with the appropriate isocyanates V R1-N═C═O.
  • Figure US20080167339A1-20080710-C00004
  • Since acids are usually liberated in these reactions, it is advisable to add bases such as pyridine, triethylamine, sodium hydroxide solution or alkali metal carbonates for expedition. The reactions can be carried out in wide temperature ranges. It has usually proved to be advantageous to operate at from 0° C. to the boiling point of the solvent used. Examples of solvents employed are methylene chloride, THF, DMF, toluene, ethyl acetate, n-heptane, dioxane, diethyl ether or pyridine. If anhydrous conditions are used, strong bases such as lithium hydride, sodium hydride or potassium tert-butoxide in aprotic solvents such as THF or DMF have also proved suitable.
  • The azolopyridin-2-one derivatives or corresponding aza-substituted derivatives employed as starting compounds II are commercially available or can be prepared by processes known from the literature (e.g. C. Flouzat, Y. Bresson, A. Mattio, J. Bonnet, G. Guillaumet J. Med. Chem. 1993, 36, 497-503; F. Mutterer, C. D. Weis, J. Het. Chem. 1976, 13, 1103-1104; K. Bowden, G. Crank, W. J. Ross, J. Chem. Soc. 1968, 172-185).
  • The examples detailed below serve to illustrate the invention without, however, restricting it.
  • EXAMPLES Example 1 2-Oxo-oxazolo[4,5-b]pyridin-3-(2-methylbenzyl)carboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were dissolved in 5 ml of THF. Addition of 129.8 mg (0.882 mmol) of 2-methylbenzyl isocyanate was followed by stirring at 70° C. for 5 h, addition once again of the same amount of 2-methylbenzyl isocyanate, and stirring 70° C. for 5 h. The reaction mixture was concentrated and purified by preparative HPLC (PR18, acetonitrile/water 0.1% TFA). Yield: 55 mg (26%), M+H+: 284.1.
  • Example 2 6-Bromo-2-oxo-oxazolo[4,5-b]pyridine-3-(2-methylbenzyl)carboxamide
  • 100 mg (0.465 mmol) of 6-bromo-3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 102.7 mg (0.698 mmol) of 2-methylbenzyl isocyanate in THF at 70° C. Yield: 13 mg (8%), M+H+: 362.05.
  • Example 3 2-Oxo-oxazolo[4,5-b]pyridine-3-hexylcarboxamide
  • 100 mg (0.74 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 102.8 mg (0.809 mmol) of 1-isocyanatohexane in dioxane at 80° C. Yield: 77 mg (40%), M+H+: 264.12.
  • Example 4 2-Oxo-oxazolo[4,5-b]pyridine-3-(3-methylbenzyl)carboxamide
  • 200 mg (1.47 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 324.3 mg (2.2 mmol) of 1-isocyanatomethyl-3-methylbenzene in dioxane at 80° C. Yield: 322 mg (77%), M+H+: 284.1.
  • Example 5 2-Oxo-oxazolo[4,5-b]pyridine-3-(3,4-dimethylbenzyl)carboxamide
  • 200 mg (1.47 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 284 mg (1.76 mmol) of 3,4-dimethylbenzyl isocyanate in dioxane at 80° C. Yield: 110 mg (25%), M+H+: 298.1.
  • Example 6 3-(4-Methylpiperidine-1-carbonyl)-3H-oxazolo[4,5-b]pyridin-2-one
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one, 235 mg (1.45 mmol) of 4-methylpiperidine-1-carbonyl chloride and 101 μl (2.18 mmol) of triethylamine were stirred in 5 ml of pyridine at room temperature for 3 h. The reaction mixture was concentrated, and the residue was dissolved in water and extracted with ethyl acetate. The organic phase was concentrated and purified by preparative HPLC (PR18, acetonitrile/water 0.1% TFA). Yield: 6 mg (3%), M+H+: 262.12.
  • Example 7 6-Bromo-3-(4-methylpiperidine-1-carbonyl)-3H-oxazolo[4,5-b]pyridin-2-one
  • 100 mg (0.46 mmol) of 6-bromo-3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 6 with 150 mg (0.93 mmol) of 4-methylpiperidin-1-carbonyl chloride. Yield: 78 mg (50%), M+H+: 340.09.
  • Example 8 2-Oxo-oxazolo[4,5-b]pyridine-3-phenethylcarboxamide
  • 300 mg (2.2 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 389 mg (2.6 mmol) of (2-isocyanatoethyl)benzene in dioxane at 80° C. Yield: 436 mg (70%), M+H+: 284.07.
  • Example 9 2-Oxo-oxazolo[4,5-b]pyridine-3-heptylcarboxamide
  • 200 mg (1.47 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 207 mg (1.47 mmol) of 1-isocyanatoheptane in dioxane at 80° C. Yield: 416 mg (10%), M+H+: 278.15.
  • Example 10 2-Oxo-oxazolo[4,5-b]pyridine-3-benzylcarboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 117 mg (0.88 mmol) of isocyanatomethylbenzene in dioxane at 80° C. Yield: 30 mg (15%), M+H+: 270.11.
  • Example 11 2-Oxo-oxazolo[4,5-b]pyridine-3-butylcarboxamide
  • 300 mg (2.2 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 262 mg (2.6 mmol) of 1-isocyanatobutane in dioxane at 80° C. Yield: 91 mg (18%), M+H+: 236.15.
  • Example 12 6-Bromo-2-oxo-oxazolo[4,5-b]pyridine-3-benzylcarboxamide
  • 100 mg (0.46 mmol) of 6-bromo-3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 74.3 mg (0.56 mmol) of isocyanatomethylbenzene in dioxane at 80° C. Yield: 10 mg (6%), M+H+: 348.10.
  • Example 13 2-Oxo-oxazolo[4,5-b]pyridin-3-(1,2,3,4-tetrahydronaphthalen-1-yl)carboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 127 mg (0.735 mmol) of 1-isocyanato-1,2,3,4-tetrahydronaphthalene in dioxane at 80° C. Yield: 29 mg (13%), M+H+: 310.10.
  • Example 14 2-Oxo-oxazolo[4,5-b]pyridine-3-(R)-indan-1-yl)carboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 140 mg (0.88 mmol) of (R)-1-isocyanatoindane in dioxane at 60° C. Yield: 22 mg (10%), M+H+: 296.14.
  • Example 15 2-Oxo-oxazolo[4,5-b]pyridine-3-(S)-indan-1-yl)carboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 140 mg (0.88 mmol) of (S)-1-isocyanatoindane in dioxane at 60° C. Yield: 8 mg (40%), M+H+: 296.14.
  • Example 16 2-Oxo-oxazolo[4,5-b]pyridin-3-dodecylcarboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 155.3 mg (0.735 mmol) of 1-isocyanatododecane in dioxane at 80° C. Yield: 33 mg (13%), M+H+: 348.25.
  • Example 17 5-Methyl-2-oxo-oxazolo[4,5-b]pyridine-3-(2-methylbenzyl)carboxamide
  • 150 mg (1 mmol) of 5-methyl-3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 176 mg (1.2 mmol) of 1-isocyanatomethyl-2-methyl-benzene and 100 μl of pyridine in toluene at 115° C. Yield: 3 mg (1%), M+H+: 298.13.
  • Example 18 2-Oxo-oxazolo[4,5-b]pyridine-3-cyclohexylmethylcarboxamide
  • 100 mg (0.735 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 102 mg (0.735 mmol) of isocyanatomethylcyclohexane in dioxane at 80° C. Yield: 43 mg (21%), M+H+: 276.13.
  • Example 19 2-Oxo-oxazolo[4,5-b]pyridine-3-octylcarboxamide
  • 200 mg (1.47 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 273.7 mg (1.76 mmol) of 1-isocyanatooctane in dioxane at 80° C. Yield: 40 mg (9%), M+H+: 292.10.
  • Example 20 2-Oxo-oxazolo[4,5-b]pyridine-3-nonylcarboxamide
  • 200 mg (1.47 mmol) of 3H-oxazolo[4,5-b]pyridin-2-one were reacted in analogy to Example 1 with 298.4 mg (1.76 mmol) of 1-isocyanatononane in dioxane at 80° C. Yield: 40 mg (9%), M+H+: 306.20.

Claims (23)

1. A compound of formula I
Figure US20080167339A1-20080710-C00005
wherein:
X is independently selected from ═C(—R)— and ═N—, with the proviso that at least one and at most two of X is ═N—;
Y is —O— or —S—;
R is independently selected from hydrogen, halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy-(C1-C3)-alkylene, hydroxy, (C1-C6)-alkylmercapto, amino, (C1-C6)-alkylamino, di-(C2-C12)-alkylamino, (C1-C6)-alkylcarbonyl, COOR3, trifluoromethyl, (C1-C6)-alkylsulfonyl, (C1-C6)-alkylsulfinyl, aminosulfonyl, pentafluorosulfanyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, CO—NR6R7, O—CO—NR6R7, O—CO—(C1-C6)-alkylene-CO—O—(C1-C6)-alkyl, O—CO—(C1-C6)-alkylene-CO—OH, O—CO—(C1-C6)-alkylene-CO—NR6R7 and unsubstituted or mono- or poly-F-substituted (C1-C6)-alkyloxy;
R1 is selected from (C4-C16)-alkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C3-C12)-cycloalkyl, and (C8-C14)-bicycle, wherein each of said aryl, heteroaryl, cycloalkyl and bicycle may be substituted one or more times by a substituent selected from halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, (C1-C6)-alkylmercapto, amino, (C1-C6)-alkylamino, di-(C2-C12)-alkylamino, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, (C1-C6)-alkoxycarbonyl, (C1-C6)-alkylcarbonyl, cyano, trifluoromethyl, trifluoromethyloxy, (C1-C6)-alkylsulfonyl, and aminosulfonyl;
R2 is selected from hydrogen and (C1-C6)-alkyl; or, in the alternative,
R1 and R2 form, together with the nitrogen atom to which they are attached, a monocyclic, saturated or partially unsaturated 4- to 7-membered ring system or a bicyclic saturated or partially unsaturated 8- to 14-membered ring system, of which individual members of the ring systems may be replaced by one to three atoms or atomic groups selected from —CHR4-, —CR4R5-, —(C═R4)-, —NR6-, —C(═O)—, —O—, —S—, —SO—, —SO2—, with the proviso that no two of the units —O—, —S—, —SO—, and —SO2— may be adjacent to each other;
R3 is hydrogen, (C1-C6)-alkyl, or benzyl;
R4 and R5 are independently selected from (C1-C6)-alkyl, halogen, trifluoromethyl, COOR3, cyclopropyl, and cyclopropylene; and
R6 and R7 are independently selected from hydrogen, (C1-C6)-alkyl, —(C6-C10)-aryl, (C5-C12)-heteroaryl, (C3-C12)-cycloalkyl, (C1-C4)-alkylene-(C6-C10)-aryl, (C1-C4)-alkylene-(C5-C12)-heteroaryl, (C1-C4)-alkylene-(C4-C12)-cycloalkyl, and (C8-C14)-bicycle;
or a tautomer or a salt thereof.
2. The compound of formula I as claimed in claim 1, wherein
X is identically or differently ═C(—R)— or ═N—, where one ═C(—R—)— is replaced by ═N—;
Y is —O— or —S—;
R is independently selected from hydrogen, halogen, (C1-C6)-alkyl, hydroxy, trifluoromethyl, mono-(C1-C6)-alkylaminocarbonyl, di-(C2-C8)-alkylaminocarbonyl, COOR3, (C1-C6)-alkylsulfonyl, aminosulfonyl, (C6-C10)-aryl, (C5-C12)-heteroaryl, (C1-C6)-alkylcarbonyl, O—CO—NR6R7, O—CO—(C1-C6)-alkylene-CO—O—(C1-C6)-alkyl, O—CO—(C1-C6)-alkylene-CO—NR6R7 and unsubstituted or mono- or poly-F-substituted (C1-C6)-alkyloxy;
R1 is selected from (C6-C12)-alkyl, (C1-C2)-alkylene-(C6-C10)-aryl, (C1-C2)-alkylene-(C5-C12)-heteroaryl, (C1-C2)-alkylene-(C4-C12)-cycloalkyl, and (C8-C14)-bicycle, wherein each of aryl, heteroaryl, cycloalkyl and bicycle may be substituted one or more times by a substituent selected from halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy, hydroxy, amino, (C1-C6)-alkylamino, and trifluoromethyl;
R2 is hydrogen; or, in the alternative,
R1 and R2 together with the nitrogen atom to which they are bonded, form a monocyclic, saturated 5- to 6-membered ring system or a bicyclic saturated or partially unsaturated 9- to 10 membered ring system, of which individual members of the ring systems may be replaced by one to three atoms or atomic groups selected from —CHR4-, —CR4R5-, —(C═R4)-, —NR6-, —O—, and —S—, with the proviso that no two —O— and —S— units may be adjacent;
R3 is hydrogen, (C1-C6)-alkyl or benzyl;
R4 and R5 are independently selected from (C1-C6)-alkyl, halogen, trifluoromethyl, COOR3, cyclopropyl, and cyclopropylene; and
R6 and R7 are independently selected from hydrogen, (C1-C6)-alkyl, (C6-C10)-aryl, (C3-C12)-cycloalkyl, and (C1-C4)-alkylene-(C6-C10)-aryl.
3. The compound of formula I as claimed in claim 1, wherein
X in position 5, 6 and 7 is independently selected from ═C(—R)—, and in position 4 X is ═N—.
4. The compound of formula I as claimed in claim 1, wherein
X in position 4 or 7 is ═C(—R)—, with R being hydrogen, and in position 5 or 6 X is ═C(—R—)— with R being other than hydrogen.
5. The compound of formula I as claimed in claim 1, in which
Y is —O— or —S—;
R1 is selected from (C6-C12)-alkyl, benzyl, (C1)-alkylene-(C5-C12)-heteroaryl, (C1)-alkylene-(C4-C12)-cycloalkyl, and (C8-C14)-bicycle, where benzyl, heteroaryl, cycloalkyl or bicycle may be substituted by a substituent selected from halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy and trifluoromethyl;
R2 is hydrogen; or, alternatively,
R1 and R2 together with the nitrogen atom bearing them, are a monocyclic, saturated 5- to 6-membered ring system, of which individual members of the ring systems may be replaced by one to two atoms or atomic groups selected from —CHR4-, and —NR6 in which R4 is as defined above, and R6 is (C1-C6)-alkyl or cyclopropyl.
6. The compound of formula I as claimed in claim 1, wherein
Y is —O—;
R is independently selected from hydrogen, halogen and (C1-C6)-alkyl;
R1 is selected from (C6-C12)-alkyl, benzyl, —CH2-alkylene-(C5-C12)-heteroaryl, —CH2—(C4-C12)-cycloalkyl, and (C8-C14)-bicycle, where benzyl, heteroaryl, cycloalkyl and bicycle may be independently substituted by halogen, (C1-C6)-alkyl, (C1-C3)-alkyloxy or trifluoromethyl; and
R2 is hydrogen.
7. The compound of formula I as claimed in claim 1, wherein
Y is —O—;
R is independently selected from hydrogen, halogen and (C1-C6)-alkyl;
R1 is selected from (C6-C12)-alkyl, benzyl, —CH2—(C4-C12)-cyclohexyl, indanyl, and tetrahydronaphthyl, where benzyl, cyclohexyl, indanyl and tetrahydronaphthyl may be independently substituted by (C1-C6)-alkyl; and
R2 is hydrogen.
8. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
9. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 2 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
10. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 3 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
11. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 4 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
12. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 5 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
13. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 6 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
14. A pharmaceutical composition comprising one or more compounds of formula I as claimed in claim 7 or a pharmaceutically acceptable salt thereof or a tautomer thereof in combination with at least one pharmaceutically acceptable excipient.
15. A method for the treatment or prevention of disorders of fatty acid metabolism and glucose utilization disorders in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
16. A method for the treatment or prevention of insulin resistance in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
17. A method for the treatment or prevention of diabetes mellitus and the sequelae associated therewith in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
18. A method for the treatment or prevention of dyslipidemias and the sequelae thereof in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
19. A method for the treatment or prevention of conditions associated with metabolic syndrome in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
20. A method for the treatment or prevention of conditions associated with reduced HDL level in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
21. A method for the treatment or prevention of atherosclerotic disorders in a patient, which comprises administering to said patient a therapeutically effective amount of a compound of formula I as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
22. A process for preparing a compound of formula I as claimed in claim 1, which comprises:
acylating an azolopyridin-2-one derivative of formula II with a carbamoyl chloride of formula III;
or
in two stages, reacting said azolopyridin-2-one derivative of formula II first with phosgene or a phosgene equivalent chosen from trichloromethyl chlorocarbonate, ditrichloromethyl carbonate or 4-nitrophenyl chloroformate, and, in a second step, reacting the resulting intermediate with an amine of formula IV,
in which the substituents are as defined in claim 1.
Figure US20080167339A1-20080710-C00006
23. A process for preparing a compound of formula I as claimed in claim 1, wherein R2 is hydrogen, which comprises reacting an azolopyridin-2-one derivative of formula II with an isocyanate of formula V: O═C═N—R1.
Figure US20080167339A1-20080710-C00007
US11/950,932 2005-06-09 2007-12-05 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors Expired - Fee Related US7846943B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/914,045 US20110039883A1 (en) 2005-06-09 2010-10-28 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DE102005026762A DE102005026762A1 (en) 2005-06-09 2005-06-09 Azolopyridin-2-one derivatives as inhibitors of lipases and phospholipases
DE102005026762 2005-06-09
DE102005026762.9 2005-06-09
PCT/EP2006/005095 WO2006131231A1 (en) 2005-06-09 2006-05-27 Azolopyridine-2-on derivatives as lipase and phospholipase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/005095 Continuation WO2006131231A1 (en) 2005-06-09 2006-05-27 Azolopyridine-2-on derivatives as lipase and phospholipase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/914,045 Continuation US20110039883A1 (en) 2005-06-09 2010-10-28 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors

Publications (2)

Publication Number Publication Date
US20080167339A1 true US20080167339A1 (en) 2008-07-10
US7846943B2 US7846943B2 (en) 2010-12-07

Family

ID=37076291

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/950,932 Expired - Fee Related US7846943B2 (en) 2005-06-09 2007-12-05 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors
US12/914,045 Abandoned US20110039883A1 (en) 2005-06-09 2010-10-28 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/914,045 Abandoned US20110039883A1 (en) 2005-06-09 2010-10-28 Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors

Country Status (30)

Country Link
US (2) US7846943B2 (en)
EP (1) EP1896485B1 (en)
JP (1) JP2008542410A (en)
KR (1) KR20080019215A (en)
CN (1) CN101193896A (en)
AR (1) AR054132A1 (en)
AT (1) ATE468342T1 (en)
AU (1) AU2006257022A1 (en)
BR (1) BRPI0611688A2 (en)
CA (1) CA2610325A1 (en)
CY (1) CY1110213T1 (en)
DE (2) DE102005026762A1 (en)
DK (1) DK1896485T3 (en)
ES (1) ES2346561T3 (en)
HR (1) HRP20100409T1 (en)
IL (1) IL187730A0 (en)
MA (1) MA29481B1 (en)
MX (1) MX2007015347A (en)
MY (1) MY142792A (en)
NO (1) NO20080133L (en)
NZ (1) NZ564063A (en)
PL (1) PL1896485T3 (en)
PT (1) PT1896485E (en)
RS (1) RS51268B (en)
RU (1) RU2414473C2 (en)
SI (1) SI1896485T1 (en)
TW (1) TW200716653A (en)
UY (1) UY29593A1 (en)
WO (1) WO2006131231A1 (en)
ZA (1) ZA200709501B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8754113B2 (en) 2009-12-15 2014-06-17 Shionogi & Co., Ltd. Oxadiazole derivative having endothelial lipase inhibitory activity
US8957219B2 (en) 2008-10-17 2015-02-17 Shionogi & Co., Ltd. Acetic acid amide derivative having inhibitory activity on endothelial lipase

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006014685A1 (en) * 2006-03-28 2007-10-04 Sanofi-Aventis New bicyclic imidazolone derivatives useful as endothelial lipase inhibitors, e.g. for treating disorders of fat metabolism or glucose utilization
EP2025674A1 (en) 2007-08-15 2009-02-18 sanofi-aventis Substituted tetra hydro naphthalines, method for their manufacture and their use as drugs
AR072707A1 (en) 2008-07-09 2010-09-15 Sanofi Aventis HETEROCICLIC COMPOUNDS, PROCESSES FOR THEIR PREPARATION, DRUGS THAT UNDERSTAND THESE COMPOUNDS AND THE USE OF THEM
WO2011059839A1 (en) * 2009-10-29 2011-05-19 Sirtris Pharmaceuticals, Inc. Bicyclic pyridines and analogs as sirtuin modulators
US8933024B2 (en) 2010-06-18 2015-01-13 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
CN114560873B (en) * 2021-12-27 2023-02-28 浙江日出药业有限公司 Preparation method of 3-chloromethyl-6-chloro-oxazole [4,5-b ] pyridine-2 (3H) ketone

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4988719A (en) * 1988-03-25 1991-01-29 Bayer Aktiengesellschaft 3,7-disubstituted benzothiazolones and fungicidal use thereof
US5147883A (en) * 1990-06-22 1992-09-15 Adir Et Compagnie Acylbenzoxazolinones compounds

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2663336B1 (en) 1990-06-18 1992-09-04 Adir NOVEL PEPTIDE DERIVATIVES, THEIR PREPARATION PROCESS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME.
FR2713225B1 (en) 1993-12-02 1996-03-01 Sanofi Sa Substituted N-piperidino-3-pyrazolecarboxamide.
IL108633A (en) 1993-02-15 1998-07-15 Wellcome Found Hypolipidaemic benzothiazepine derivatives their preparation and pharmaceutical compositions containing them
IL108634A0 (en) 1993-02-15 1994-05-30 Wellcome Found Hypolipidaemic heterocyclic compounds, their prepatation and pharmaceutical compositions containing them
JP3144624B2 (en) 1995-06-02 2001-03-12 杏林製薬株式会社 N-benzyldioxothiazolidylbenzamide derivative and method for producing the same
AU727775B2 (en) 1996-01-17 2000-12-21 Novo Nordisk A/S Fused 1,2,4-thiadiazine and fused 1,4-thiazine derivatives, their preparation and use
PT944648E (en) 1996-08-30 2007-06-26 Novo Nordisk As Glp-1 derivatives
TW492957B (en) 1996-11-07 2002-07-01 Novartis Ag N-substituted 2-cyanopyrrolidnes
EP0958296B1 (en) 1996-12-31 2003-07-30 Dr. Reddy's Laboratories Ltd. Heterocyclic compounds process for their preparation and pharmaceutical compositions containing them and their use in the treatment of diabetes and related diseases
DE19726167B4 (en) 1997-06-20 2008-01-24 Sanofi-Aventis Deutschland Gmbh Insulin, process for its preparation and pharmaceutical preparation containing it
BR9810592A (en) 1997-07-16 2000-09-12 Novo Nordisk As Compound, processes for preparing a compound, for treating or preventing diseases of the endocrine system and for the manufacture of a drug, pharmaceutical composition, and, use of a compound
CO4970713A1 (en) 1997-09-19 2000-11-07 Sanofi Synthelabo DERIVATIVES OF CARBOXAMIDOTIAZOLES, THEIR PREPARATION, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM
DE19823831A1 (en) 1998-05-28 1999-12-02 Probiodrug Ges Fuer Arzneim New pharmaceutical use of isoleucyl thiazolidide and its salts
SE9801992D0 (en) 1998-06-04 1998-06-04 Astra Ab New 3-aryl-2-hydroxypropionic acid derivative I
MA26634A1 (en) 1998-06-04 2004-12-20 Astra Ab NOVEL 3-ARYL PROPIONIC ACID DERIVATIVES AND THE LIKE
US6221897B1 (en) 1998-06-10 2001-04-24 Aventis Pharma Deutschland Gmbh Benzothiepine 1,1-dioxide derivatives, a process for their preparation, pharmaceuticals comprising these compounds, and their use
DE19828113A1 (en) 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
DE19828114A1 (en) 1998-06-24 2000-01-27 Probiodrug Ges Fuer Arzneim Produgs of unstable inhibitors of dipeptidyl peptidase IV
DE19845405C2 (en) 1998-10-02 2000-07-13 Aventis Pharma Gmbh Aryl-substituted propanolamine derivatives and their use
GB9900416D0 (en) 1999-01-08 1999-02-24 Alizyme Therapeutics Ltd Inhibitors
DE19916108C1 (en) 1999-04-09 2001-01-11 Aventis Pharma Gmbh 1,4-Benzothiazepine-1,1-dioxide derivatives substituted with sugar residues, process for their preparation and their use
EP1173438A1 (en) 1999-04-16 2002-01-23 Novo Nordisk A/S Substituted imidazoles, their preparation and use
PT1177176E (en) 1999-04-28 2006-08-31 Aventis Pharma Gmbh DERIVATIVES OF TRIARILIC ACIDS AS LIGANDS OF PPAR RECEPTORS.
SK15532001A3 (en) 1999-04-28 2002-06-04 Aventis Pharma Deutschland Gmbh Di-aryl acid derivatives as ppar receptor ligands
AU4808300A (en) 1999-04-30 2000-11-17 Neurogen Corporation 9h-pyrimido(4,5-b)indole derivatives: crf1 specific ligands
GB9911863D0 (en) 1999-05-21 1999-07-21 Knoll Ag Therapeutic agents
CA2376919C (en) 1999-06-18 2008-11-04 Merck & Co., Inc. Arylthiazolidinedione and aryloxazolidinedione derivatives
WO2001004146A2 (en) 1999-07-09 2001-01-18 Cohesion Technologies, Inc. Ecarin polypeptides, polynucleotides encoding ecarin, and methods for use thereof
DK1204654T3 (en) 1999-07-29 2003-11-17 Lilly Co Eli Benzofurylpiperazines and benzofurylhomopiperazines: Serotonin agonists
DK1218341T3 (en) 1999-09-01 2005-12-12 Sanofi Aventis Deutschland Sulfonylcarboxamide derivatives, process for their preparation and their use as a drug
TW200528436A (en) 1999-09-22 2005-09-01 Bristol Myers Squibb Co Substituted acid derivatives useful as antiodiabetic and antiobesity agents and method
SE9904413D0 (en) 1999-12-03 1999-12-03 Astra Ab Comminuted form
EE200200278A (en) 1999-12-03 2003-06-16 Astrazeneca Ab Crystalline form of (S) -2-ethoxy-3- [4- (2- {4-methanesulfonyloxyphenyl} ethoxy) phenyl] propanoic acid
CA2400226C (en) 2000-03-31 2007-01-02 Probiodrug Ag Method for the improvement of islet signaling in diabetes mellitus and for its prevention
WO2001081327A1 (en) 2000-04-25 2001-11-01 Kyorin Pharmaceutical Co., Ltd. Novel stable crystal of thiazolidinedione derivative and process for producing the same
CA2407538C (en) 2000-04-28 2007-01-09 Asahi Kasei Kabushiki Kaisha Novel bicyclic compounds
EP1280777B1 (en) 2000-05-11 2005-11-23 Bristol-Myers Squibb Company Tetrahydroisoquinoline analogs useful as growth hormone secretagogues
AU2001264977B2 (en) 2000-05-30 2005-04-14 Merck & Co., Inc. Melanocortin receptor agonists
DE50102147D1 (en) 2000-06-09 2004-06-03 Aventis Pharma Gmbh ACYLPHENYL URINE DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A MEDICINAL PRODUCT
TWI243162B (en) 2000-11-10 2005-11-11 Taisho Pharmaceutical Co Ltd Cyanopyrrolidine derivatives
AU1609702A (en) 2000-12-21 2002-07-01 Aventis Pharma Gmbh Novel 1,2-diphenzylazetidinones, method for producing the same, medicaments containing said compounds, and the use thereof for treating disorders of the lipid metabolism
PE20021091A1 (en) 2001-05-25 2003-02-04 Aventis Pharma Gmbh DERIVATIVES OF PHENYLUREA SUBSTITUTED WITH CARBONAMIDE AND PROCEDURE FOR THEIR PREPARATION
DE10154689A1 (en) 2001-11-09 2003-05-22 Probiodrug Ag Substituted amino ketone compounds
EP1275653A1 (en) 2001-07-10 2003-01-15 Bayer CropScience S.A. Oxazolopyridines and their use as fungicides
KR100915108B1 (en) 2001-08-31 2009-09-03 사노피-아벤티스 도이칠란트 게엠베하 Diarylcycloalkyl derivatives as PPAR activators and a pharmaceutical composition comprising the same
KR20040068240A (en) * 2001-12-14 2004-07-30 노보 노르디스크 에이/에스 Compounds and uses thereof for decreasing activity of hormone-sensitive lipase
IL164249A0 (en) 2002-04-11 2005-12-18 Aventis Pharma Gmbh Acyl-3-carboxphenylurea derivatives, processes forpreparing them and their use
DE10215908B4 (en) 2002-04-11 2005-08-18 Aventis Pharma Deutschland Gmbh Acyl-3-carboxyphenyl-urea derivatives and their use as medicaments
DE10225635C1 (en) 2002-06-07 2003-12-24 Aventis Pharma Gmbh N-benzoylureido-cinnamic acid derivatives, process for their preparation and their use
DE10231370B4 (en) 2002-07-11 2006-04-06 Sanofi-Aventis Deutschland Gmbh Thiophene glycoside derivatives, medicaments containing these compounds and methods of making these medicaments
DE10247680B4 (en) 2002-10-12 2005-09-01 Aventis Pharma Deutschland Gmbh New bicyclic inhibitors of the hormone sensitive lipase
DE10258008B4 (en) 2002-12-12 2006-02-02 Sanofi-Aventis Deutschland Gmbh Heterocyclic fluoroglycoside derivatives, medicaments containing these compounds and methods of making these medicaments
DE10258007B4 (en) 2002-12-12 2006-02-09 Sanofi-Aventis Deutschland Gmbh Aromatic fluoroglycoside derivatives, medicaments containing these compounds and methods for the preparation of these medicaments
US7595403B2 (en) 2003-04-01 2009-09-29 Eli Lilly And Company Benzisothiazol-3-one-carboxylic acid amides as phospholipase inhibitors
EP1613608A1 (en) 2003-04-01 2006-01-11 Eli Lilly And Company Phospholipase inhibitors
DE102004005172A1 (en) 2004-02-02 2005-08-18 Aventis Pharma Deutschland Gmbh Indazole derivatives as inhibitors of the hormone sensitive lipase
DE102004028241B4 (en) 2004-06-11 2007-09-13 Sanofi-Aventis Deutschland Gmbh New fluoroglycoside derivatives of pyrazoles, medicines containing these compounds and manufacture of these medicines

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4988719A (en) * 1988-03-25 1991-01-29 Bayer Aktiengesellschaft 3,7-disubstituted benzothiazolones and fungicidal use thereof
US5147883A (en) * 1990-06-22 1992-09-15 Adir Et Compagnie Acylbenzoxazolinones compounds

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957219B2 (en) 2008-10-17 2015-02-17 Shionogi & Co., Ltd. Acetic acid amide derivative having inhibitory activity on endothelial lipase
US8754113B2 (en) 2009-12-15 2014-06-17 Shionogi & Co., Ltd. Oxadiazole derivative having endothelial lipase inhibitory activity

Also Published As

Publication number Publication date
PL1896485T3 (en) 2010-10-29
PT1896485E (en) 2010-08-05
US7846943B2 (en) 2010-12-07
US20110039883A1 (en) 2011-02-17
JP2008542410A (en) 2008-11-27
UY29593A1 (en) 2007-01-31
EP1896485B1 (en) 2010-05-19
WO2006131231A1 (en) 2006-12-14
ZA200709501B (en) 2008-10-29
ES2346561T3 (en) 2010-10-18
SI1896485T1 (en) 2010-09-30
IL187730A0 (en) 2008-08-07
MY142792A (en) 2010-12-31
DE502006006986D1 (en) 2010-07-01
HRP20100409T1 (en) 2010-09-30
RS51268B (en) 2010-12-31
CY1110213T1 (en) 2012-05-23
MX2007015347A (en) 2008-02-15
MA29481B1 (en) 2008-05-02
BRPI0611688A2 (en) 2010-09-28
RU2414473C2 (en) 2011-03-20
EP1896485A1 (en) 2008-03-12
CN101193896A (en) 2008-06-04
TW200716653A (en) 2007-05-01
NZ564063A (en) 2010-07-30
RU2007145499A (en) 2009-06-20
AU2006257022A1 (en) 2006-12-14
CA2610325A1 (en) 2006-12-14
AR054132A1 (en) 2007-06-06
KR20080019215A (en) 2008-03-03
ATE468342T1 (en) 2010-06-15
NO20080133L (en) 2008-03-05
DE102005026762A1 (en) 2006-12-21
DK1896485T3 (en) 2010-09-06

Similar Documents

Publication Publication Date Title
US8921404B2 (en) Azole derivatives in the form of lipase and phospholipase inhibitors
US7968719B2 (en) Indazole derivatives as inhibitors of hormone sensitive lipase
US7846943B2 (en) Azolopyridin-2-one derivatives as lipase and phospholipase inhibitors
US7709513B2 (en) Benzooxazol-2-one derivatives as lipase and phospholipase inhibitors
DE102005049954A1 (en) Triazolopyridine derivatives as inhibitors of lipases and phospholipases
US8008331B2 (en) Benzothiazol-2-one derivatives as lipase and phospholipase inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANOFI-AVENTIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZOLLER, GERHARD;PETRY, STEFAN;MULLER, GUNTER;AND OTHERS;REEL/FRAME:020678/0684;SIGNING DATES FROM 20080208 TO 20080215

Owner name: SANOFI-AVENTIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZOLLER, GERHARD;PETRY, STEFAN;MULLER, GUNTER;AND OTHERS;SIGNING DATES FROM 20080208 TO 20080215;REEL/FRAME:020678/0684

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: SANOFI, FRANCE

Free format text: CHANGE OF NAME;ASSIGNOR:SANOFI-AVENTIS;REEL/FRAME:028413/0927

Effective date: 20110511

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552)

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20221207